101
|
Krakovski MA, Arora N, Jain S, Glover J, Dombrowski K, Hernandez B, Yadav H, Sarma AK. Diet-microbiome-gut-brain nexus in acute and chronic brain injury. Front Neurosci 2022; 16:1002266. [PMID: 36188471 PMCID: PMC9523267 DOI: 10.3389/fnins.2022.1002266] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
In recent years, appreciation for the gut microbiome and its relationship to human health has emerged as a facilitator of maintaining healthy physiology and a contributor to numerous human diseases. The contribution of the microbiome in modulating the gut-brain axis has gained significant attention in recent years, extensively studied in chronic brain injuries such as Epilepsy and Alzheimer’s Disease. Furthermore, there is growing evidence that gut microbiome also contributes to acute brain injuries like stroke(s) and traumatic brain injury. Microbiome-gut-brain communications are bidirectional and involve metabolite production and modulation of immune and neuronal functions. The microbiome plays two distinct roles: it beneficially modulates immune system and neuronal functions; however, abnormalities in the host’s microbiome also exacerbates neuronal damage or delays the recovery from acute injuries. After brain injury, several inflammatory changes, such as the necrosis and apoptosis of neuronal tissue, propagates downward inflammatory signals to disrupt the microbiome homeostasis; however, microbiome dysbiosis impacts the upward signaling to the brain and interferes with recovery in neuronal functions and brain health. Diet is a superlative modulator of microbiome and is known to impact the gut-brain axis, including its influence on acute and neuronal injuries. In this review, we discussed the differential microbiome changes in both acute and chronic brain injuries, as well as the therapeutic importance of modulation by diets and probiotics. We emphasize the mechanistic studies based on animal models and their translational or clinical relationship by reviewing human studies.
Collapse
Affiliation(s)
| | - Niraj Arora
- Department of Neurology, University of Missouri, Columbia, MO, United States
| | - Shalini Jain
- Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, FL, United States
| | - Jennifer Glover
- Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, FL, United States
| | - Keith Dombrowski
- Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, FL, United States
| | - Beverly Hernandez
- Clinical Nutrition Services, Tampa General Hospital, Tampa, FL, United States
| | - Hariom Yadav
- Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, FL, United States
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida, Tampa, FL, United States
- *Correspondence: Hariom Yadav,
| | - Anand Karthik Sarma
- Wake Forest University School of Medicine, Winston-Salem, NC, United States
- Department of Neurology, Atrium Health Wake Forest Baptist, Winston-Salem, NC, United States
- Anand Karthik Sarma,
| |
Collapse
|
102
|
Song L, Yang YT, Guo Q, Zhao XM. Cellular transcriptional alterations of peripheral blood in Alzheimer's disease. BMC Med 2022; 20:266. [PMID: 36031604 PMCID: PMC9422129 DOI: 10.1186/s12916-022-02472-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 07/11/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Alzheimer's disease (AD), a progressive neurodegenerative disease, is the most common cause of dementia worldwide. Accumulating data support the contributions of the peripheral immune system in AD pathogenesis. However, there is a lack of comprehensive understanding about the molecular characteristics of peripheral immune cells in AD. METHODS To explore the alterations of cellular composition and the alterations of intrinsic expression of individual cell types in peripheral blood, we performed cellular deconvolution in a large-scale bulk blood expression cohort and identified cell-intrinsic differentially expressed genes in individual cell types with adjusting for cellular proportion. RESULTS We detected a significant increase and decrease in the proportion of neutrophils and B lymphocytes in AD blood, respectively, which had a robust replicability across other three AD cohorts, as well as using alternative algorithms. The differentially expressed genes in AD neutrophils were enriched for some AD-associated pathways, such as ATP metabolic process and mitochondrion organization. We also found a significant enrichment of protein-protein interaction network modules of leukocyte cell-cell activation, mitochondrion organization, and cytokine-mediated signaling pathway in neutrophils for AD risk genes including CD33 and IL1B. Both changes in cellular composition and expression levels of specific genes were significantly associated with the clinical and pathological alterations. A similar pattern of perturbations on the cellular proportion and gene expression levels of neutrophils could be also observed in mild cognitive impairment (MCI). Moreover, we noticed an elevation of neutrophil abundance in the AD brains. CONCLUSIONS We revealed the landscape of molecular perturbations at the cellular level for AD. These alterations highlight the putative roles of neutrophils in AD pathobiology.
Collapse
Affiliation(s)
- Liting Song
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, 200433, China
| | - Yucheng T Yang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, 200433, China.,MOE Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200433, China.,Zhangjiang Fudan International Innovation Center, Shanghai, 200433, China
| | - Qihao Guo
- Department of Gerontology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | | | - Xing-Ming Zhao
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, 200433, China. .,MOE Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200433, China. .,Zhangjiang Fudan International Innovation Center, Shanghai, 200433, China. .,International Human Phenome Institutes (Shanghai), Shanghai, 200433, China.
| |
Collapse
|
103
|
Sex and APOE Genotype Alter the Basal and Induced Inflammatory States of Primary Microglia from APOE Targeted Replacement Mice. Int J Mol Sci 2022; 23:ijms23179829. [PMID: 36077227 PMCID: PMC9456163 DOI: 10.3390/ijms23179829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/17/2022] [Accepted: 08/19/2022] [Indexed: 11/16/2022] Open
Abstract
The sex and APOE4 genotype are significant risk factors for Alzheimer’s disease (AD); however, the mechanism(s) responsible for this interaction are still a matter of debate. Here, we assess the responses of mixed-sex and sex-specific APOE3 and APOE4 primary microglia (PMG) to lipopolysaccharide and interferon-gamma. In our investigation, inflammatory cytokine profiles were assessed by qPCR and multiplex ELISA assays. Mixed-sex APOE4 PMG exhibited higher basal mRNA expression and secreted levels of TNFa and IL1b. In sex-specific cultures, basal expression and secreted levels of IL1b, TNFa, IL6, and NOS2 were 2−3 fold higher in APOE4 female PMG compared to APOE4 males, with both higher than APOE3 cells. Following an inflammatory stimulus, the expression of pro-inflammatory cytokines and the secreted cytokine level were upregulated in the order E4 female > E4 male > E3 female > E3 male in sex-specific cultures. These data indicate that the APOE4 genotype and female sex together contribute to a greater inflammatory response in PMG isolated from targeted replacement humanized APOE mice. These data are consistent with clinical data and indicate that sex-specific PMG may provide a platform for exploring mechanisms of genotype and sex differences in AD related to neuroinflammation and neurodegeneration.
Collapse
|
104
|
Kanagasingam S, von Ruhland C, Welbury R, Singhrao SK. Antimicrobial, Polarizing Light, and Paired Helical Filament Properties of Fragmented Tau Peptides of Selected Putative Gingipains. J Alzheimers Dis 2022; 89:1279-1291. [PMID: 36031895 DOI: 10.3233/jad-220486] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Tau is an established substrate for gingipains secreted by Porphyromonas gingivalis. Hyperphosphorylation of tau and neurofibrillary tangle (NFT) formation is a defining lesion of Alzheimer's disease (AD) where NFT distribution is related to Braak stage and disease severity. OBJECTIVE To assess gingipains'-fragmented tau peptides for their antimicrobial properties and for the likelihood of paired helical/straight filament (PHF/SF) formation with implications for the NFT lesion. METHODS Seven non-phosphorylated (A-G) and three phosphorylated (A-C) tau peptides, were tested for antimicrobial properties against P. gingivalis. Polarizing light properties were determined using Congo Red staining. Secondary and tertiary structures of peptides B-F were determined using transmission electron microscopy (TEM) and circular dichroism (CD) was undertaken for the soluble peptides A in phosphorylated and non-phosphorylated states. RESULTS Phosphorylated tau peptide A displayed a significant effect against planktonic P. gingivalis. The CD results demonstrated that both peptides A, in phosphorylated and non-phosphorylated states, in aqueous solution, adopted mainly β-type structures. Non-phosphorylated peptides B-F and phosphorylated peptides B-C were insoluble and fibrillar under the TEM. The secondary and tertiary structures of the non-phosphorylated peptide B demonstrated fewer helical twists, whereas peptide C displayed significantly more helical twists along the whole fiber(s) length following its phosphorylation. CONCLUSION Phosphorylated peptide A reduced P. gingivalis viability. CD spectroscopy demonstrated the phosphorylated and the non-phosphorylated peptide A predominantly formed from β-sheet structures in aqueous solution with potential antimicrobial activity. Phosphorylation of tau peptides physically changed their tertiary structure into PHFs with potential for self-aggregation and binding to the NFT lesion.
Collapse
Affiliation(s)
- Shalini Kanagasingam
- Brain and Behavior Centre, Faculty of Clinical and Biomedical Sciences, School of Dentistry, University of Central Lancashire, Preston, UK
| | - Christopher von Ruhland
- Electron and Light Microscopy Facility, College of Biomedical and Life Sciences, Cardiff University, Wales, UK
| | - Richard Welbury
- Brain and Behavior Centre, Faculty of Clinical and Biomedical Sciences, School of Dentistry, University of Central Lancashire, Preston, UK
| | - Sim K Singhrao
- Brain and Behavior Centre, Faculty of Clinical and Biomedical Sciences, School of Dentistry, University of Central Lancashire, Preston, UK
| |
Collapse
|
105
|
Elwakil BH, Bakr BA, Aljeldah MM, Shehata NS, Shahin YH, Olama ZA, Augustyniak M, Aboul-Soud MAM, El Wakil A. Memory Impairment, Pro-Inflammatory Host Response and Brain Histopathologic Severity in Rats Infected with K. pneumoniae or P. aeruginosa Meningitis. Pathogens 2022; 11:933. [PMID: 36015052 PMCID: PMC9416464 DOI: 10.3390/pathogens11080933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 07/29/2022] [Accepted: 08/02/2022] [Indexed: 11/25/2022] Open
Abstract
Meningitis caused by Klebsiella pneumoniae and Pseudomonas aeruginosa has lately become a prevalent cause of the central nervous system (CNS) infection. Bacterial invasion into the subarachnoid space prompts the releasing mechanism of chemokines and pro-inflammatory cytokines. The present study aimed to compare K. pneumoniae and P. aeruginosa meningitis concerning the memory, pro-inflammatory mediators and brain histopathological changes at different time intervals in adult Albino rats. The animals were sacrificed at three time intervals comprising 5, 10 and 15 days after meningitis induction. Cerebrospinal fluid (CSF) culture, relative brain weights, complete blood analysis, biochemical markers, levels of cytokine, chemokine and brain-derived neurotrophic factor (BDNF), neurotransmitter acetylcholine esterase (AChE) activity, and the brain histopathology of the infected rats in comparison to those in the control group were assessed. There was a significant increase in the levels of pro-inflammatory cytokines and chemokines including TNF-α, IL-1β, IL-6 and AChE after 5 days of bacterial meningitis infection with both K. pneumoniae and P. aeruginosa. The histopathological analysis of the cerebral cortex in the P. aeruginosa meningitis model at different time intervals revealed abundant numbers of dilated and congested blood vessels with severe hemorrhage, cerebral infarct, intracellular and extracellular vacuoles, and gliosis. Fifteen days post infection, a significant reduction in the brain tissue weight was observed. The meningitis model employing P. aeruginosa exhibited more evident time-dependent severity compared to K. pneumoniae, which may advocate its validity as a simple and effective research model to study meningitis of the CNS. This model may be utilized for further investigation to ascertain the molecular and biological association between bacterial meningitis and the development of the pathophysiological hallmarks underlying Alzheimer's disease in preclinical and clinical setups. Clinical extrapolation based on studies employing animal disease models should be carefully interpreted.
Collapse
Affiliation(s)
- Bassma H. Elwakil
- Department of Medical Laboratory Technology, Faculty of Applied Health Sciences Technology, Pharos University in Alexandria, Alexandria P.O. Box 21311, Egypt
| | - Basant A. Bakr
- Department of Zoology, Faculty of Science, Alexandria University, Alexandria P.O. Box 21568, Egypt
| | - Mohammed M. Aljeldah
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, University of Hafr Al Batin, Hafr Al Batin 39524, Saudi Arabia
| | - Nourhan S. Shehata
- Department of Medical Laboratory Technology, Faculty of Applied Health Sciences Technology, Pharos University in Alexandria, Alexandria P.O. Box 21311, Egypt
| | - Yahya H. Shahin
- Department of Medical Laboratory Technology, Faculty of Applied Health Sciences Technology, Pharos University in Alexandria, Alexandria P.O. Box 21311, Egypt
| | - Zakia A. Olama
- Department of Botany and Microbiology, Faculty of Science, Alexandria University, Alexandria P.O. Box 21568, Egypt
| | - Maria Augustyniak
- Faculty of Natural Sciences, Institute of Biology, Biotechnology and Environmental Protection, University of Silesia in Katowice, Bankowa 9, 40-007 Katowice, Poland
| | - Mourad A. M. Aboul-Soud
- Chair of Medical and Molecular Genetics Research, Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, P.O. Box 10219, Riyadh 11433, Saudi Arabia
| | - Abeer El Wakil
- Department of Biological and Geological Sciences, Faculty of Education, Alexandria University, Alexandria P.O. Box 21526, Egypt
| |
Collapse
|
106
|
Sun E, Motolani A, Campos L, Lu T. The Pivotal Role of NF-kB in the Pathogenesis and Therapeutics of Alzheimer's Disease. Int J Mol Sci 2022; 23:8972. [PMID: 36012242 PMCID: PMC9408758 DOI: 10.3390/ijms23168972] [Citation(s) in RCA: 136] [Impact Index Per Article: 45.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 08/05/2022] [Accepted: 08/09/2022] [Indexed: 02/06/2023] Open
Abstract
Alzheimer's Disease (AD) is the most common neurodegenerative disease worldwide, with a high prevalence that is expected to double every 20 years. Besides the formation of Aβ plaques and neurofibrillary tangles, neuroinflammation is one the major phenotypes that worsens AD progression. Indeed, the nuclear factor-κB (NF-κB) is a well-established inflammatory transcription factor that fuels neurodegeneration. Thus, in this review, we provide an overview of the NF-κB role in the pathogenesis of AD, including its interaction with various molecular factors in AD mice models, neurons, and glial cells. Some of these cell types and molecules include reactive microglia and astrocytes, β-secretase, APOE, glutamate, miRNA, and tau protein, among others. Due to the multifactorial nature of AD development and the failure of many drugs designed to dampen AD progression, the pursuit of novel targets for AD therapeutics, including the NF-κB signaling pathway, is rising. Herein, we provide a synopsis of the drug development landscape for AD treatment, offering the perspective that NF-κB inhibitors may generate widespread interest in AD research in the future. Ultimately, the additional investigation of compounds and small molecules that target NF-κB signaling and the complete understanding of NF-κB mechanistic activation in different cell types will broaden and provide more therapeutic options for AD patients.
Collapse
Affiliation(s)
- Emily Sun
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Biological Sciences, Columbia University, 70 Morning Drive, New York, NY 10027, USA
| | - Aishat Motolani
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Leonardo Campos
- Department of Biological Sciences, Columbia University, 70 Morning Drive, New York, NY 10027, USA
| | - Tao Lu
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
107
|
Handa T, Sasaki H, Takao M, Tano M, Uchida Y. Proteomics-based investigation of cerebrovascular molecular mechanisms in cerebral amyloid angiopathy by the FFPE-LMD-PCT-SWATH method. Fluids Barriers CNS 2022; 19:56. [PMID: 35778717 PMCID: PMC9250250 DOI: 10.1186/s12987-022-00351-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 06/21/2022] [Indexed: 11/25/2022] Open
Abstract
Background Cerebral amyloid angiopathy (CAA) occurs in 80% of patients with Alzheimer’s disease (AD) and is mainly caused by the abnormal deposition of Aβ in the walls of cerebral blood vessels. Cerebrovascular molecular mechanisms in CAA were investigated by using comprehensive and accurate quantitative proteomics. Methods Concerning the molecular mechanisms specific to CAA, formalin-fixed paraffin-embedded (FFPE) sections were prepared from patients having AD neuropathologic change (ADNC) with severe cortical Aβ vascular deposition (ADNC +/CAA +), and from patients having ADNC without vascular deposition of Aβ (ADNC +/CAA −; so called, AD). Cerebral cortical vessels were isolated from FFPE sections using laser microdissection (LMD), processed by pressure cycling technology (PCT), and applied to SWATH (sequential window acquisition of all theoretical fragment ion spectra) proteomics. Results The protein expression levels of 17 proteins in ADNC +/CAA +/H donors (ADNC +/CAA + donors with highly abundant Aβ in capillaries) were significantly different from those in ADNC +/CAA − and ADNC −/CAA − donors. Furthermore, we identified 56 proteins showing more than a 1.5-fold difference in average expression levels between ADNC +/CAA + and ADNC −/CAA − donors, and were significantly correlated with the levels of Aβ or Collagen alpha-2(VI) chain (COL6A2) (CAA markers) in 11 donors (6 ADNC +/CAA + and 5 ADNC −/CAA −). Over 70% of the 56 proteins showed ADNC +/CAA + specific changes in protein expression. The comparative analysis with brain parenchyma showed that more than 90% of the 56 proteins were vascular-specific pathological changes. A literature-based pathway analysis showed that 42 proteins are associated with fibrosis, oxidative stress and apoptosis. This included the increased expression of Heat shock protein HSP 90-alpha, CD44 antigen and Carbonic anhydrase 1 which are inhibited by potential drugs against CAA. Conclusions The combination of LMD-based isolation of vessels from FFPE sections, PCT-assisted sample processing and SWATH analysis (FFPE-LMD-PCT-SWATH method) revealed for the first time the changes in the expression of many proteins that are involved in fibrosis, ROS production and cell death in ADNC +/CAA + (CAA patients) vessels. The findings reported herein would be useful for developing a better understanding of the pathology of CAA and for promoting the discovery and development of drugs and biomarkers for CAA. Supplementary Information The online version contains supplementary material available at 10.1186/s12987-022-00351-x.
Collapse
Affiliation(s)
- Takumi Handa
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Hayate Sasaki
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Masaki Takao
- Department of Neurology and Brain Bank, Mihara Memorial Hospital, Isesaki, Japan.,Department of Clinical Laboratory, National Center of Neurology and Psychiatry, National Center Hospital, Kodaira, Japan
| | - Mitsutoshi Tano
- Department of Neurology and Brain Bank, Mihara Memorial Hospital, Isesaki, Japan
| | - Yasuo Uchida
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan. .,Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai, 980-8578, Japan.
| |
Collapse
|
108
|
Needham H, Torpey G, Flores CC, Davis CJ, Vanderheyden WM, Gerstner JR. A Dichotomous Role for FABP7 in Sleep and Alzheimer's Disease Pathogenesis: A Hypothesis. Front Neurosci 2022; 16:798994. [PMID: 35844236 PMCID: PMC9280343 DOI: 10.3389/fnins.2022.798994] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 05/10/2022] [Indexed: 11/15/2022] Open
Abstract
Fatty acid binding proteins (FABPs) are a family of intracellular lipid chaperone proteins known to play critical roles in the regulation of fatty acid uptake and transport as well as gene expression. Brain-type fatty acid binding protein (FABP7) is enriched in astrocytes and has been implicated in sleep/wake regulation and neurodegenerative diseases; however, the precise mechanisms underlying the role of FABP7 in these biological processes remain unclear. FABP7 binds to both arachidonic acid (AA) and docosahexaenoic acid (DHA), resulting in discrete physiological responses. Here, we propose a dichotomous role for FABP7 in which ligand type determines the subcellular translocation of fatty acids, either promoting wakefulness aligned with Alzheimer's pathogenesis or promoting sleep with concomitant activation of anti-inflammatory pathways and neuroprotection. We hypothesize that FABP7-mediated translocation of AA to the endoplasmic reticulum of astrocytes increases astrogliosis, impedes glutamatergic uptake, and enhances wakefulness and inflammatory pathways via COX-2 dependent generation of pro-inflammatory prostaglandins. Conversely, we propose that FABP7-mediated translocation of DHA to the nucleus stabilizes astrocyte-neuron lactate shuttle dynamics, preserves glutamatergic uptake, and promotes sleep by activating anti-inflammatory pathways through the peroxisome proliferator-activated receptor-γ transcriptional cascade. Importantly, this model generates several testable hypotheses applicable to other neurodegenerative diseases, including amyotrophic lateral sclerosis and Parkinson's disease.
Collapse
Affiliation(s)
- Hope Needham
- Department of Biology, Gonzaga University, Spokane, WA, United States
| | - Grace Torpey
- Department of Biology, Gonzaga University, Spokane, WA, United States
| | - Carlos C. Flores
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| | - Christopher J. Davis
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
- Sleep and Performance Research Center, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| | - William M. Vanderheyden
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
- Sleep and Performance Research Center, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| | - Jason R. Gerstner
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
- Sleep and Performance Research Center, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
- Steve Gleason Institute for Neuroscience, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| |
Collapse
|
109
|
Lukiw WJ. NF-kB (p50/p65)-Mediated Pro-Inflammatory microRNA (miRNA) Signaling in Alzheimer's Disease (AD). Front Mol Neurosci 2022; 15:943492. [PMID: 35836546 PMCID: PMC9274251 DOI: 10.3389/fnmol.2022.943492] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 05/27/2022] [Indexed: 11/13/2022] Open
Affiliation(s)
- Walter J. Lukiw
- LSU Neuroscience Center, Louisiana State University Health Science Center, New Orleans, LA, United States
- Department of Ophthalmology, Louisiana State University Health Science Center, New Orleans, LA, United States
- Department Neurology, Louisiana State University Health Science Center, New Orleans, LA, United States
- *Correspondence: Walter J. Lukiw
| |
Collapse
|
110
|
Pogue AI, Jaber VR, Sharfman NM, Zhao Y, Lukiw WJ. Downregulation of Neurofilament Light Chain Expression in Human Neuronal-Glial Cell Co-Cultures by a Microbiome-Derived Lipopolysaccharide-Induced miRNA-30b-5p. Front Neurol 2022; 13:900048. [PMID: 35812116 PMCID: PMC9263091 DOI: 10.3389/fneur.2022.900048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 05/12/2022] [Indexed: 11/26/2022] Open
Abstract
Microbiome-derived Gram-negative bacterial lipopolysaccharide (LPS) has been shown by multiple laboratories to reside within Alzheimer's disease (AD)-affected neocortical and hippocampal neurons. LPS and other pro-inflammatory stressors strongly induce a defined set of NF-kB (p50/p65)-sensitive human microRNAs, including a brain-enriched Homo sapien microRNA-30b-5p (hsa-miRNA-30b-5p; miRNA-30b). Here we provide evidence that this neuropathology-associated miRNA, known to be upregulated in AD brain and LPS-stressed human neuronal-glial (HNG) cells in primary culture targets the neurofilament light (NF-L) chain mRNA 3'-untranslated region (3'-UTR), which is conducive to the post-transcriptional downregulation of NF-L expression observed within both AD and LPS-treated HNG cells. A deficiency of NF-L is associated with consequent atrophy of the neuronal cytoskeleton and the disruption of synaptic organization. Interestingly, miRNA-30b has previously been shown to be highly expressed in amyloid-beta (Aβ) peptide-treated animal and cell models, and Aβ peptides promote LPS entry into neurons. Increased miRNA-30b expression induces neuronal injury, neuron loss, neuronal inflammation, impairment of synaptic transmission, and synaptic failure in neurodegenerative disease and transgenic murine models. This gut microbiota-derived LPS-NF-kB-miRNA-30b-NF-L pathological signaling network: (i) underscores a positive pathological link between the LPS of gastrointestinal (GI)-tract microbes and the inflammatory neuropathology, disordered cytoskeleton, and disrupted synaptic signaling of the AD brain and stressed brain cells; and (ii) is the first example of a microbiome-derived neurotoxic glycolipid having significant detrimental miRNA-30b-mediated actions on the expression of NF-L, an abundant neuron-specific filament protein known to be important in the maintenance of neuronal cell shape, axonal caliber, and synaptic homeostasis.
Collapse
Affiliation(s)
| | - Vivian R. Jaber
- LSU Neuroscience Center, Louisiana State University Health Science Center, New Orleans, LA, United States
| | - Nathan M. Sharfman
- LSU Neuroscience Center, Louisiana State University Health Science Center, New Orleans, LA, United States
| | - Yuhai Zhao
- LSU Neuroscience Center, Louisiana State University Health Science Center, New Orleans, LA, United States
- Department of Cell Biology and Anatomy, Louisiana State University Health Science Center, New Orleans, LA, United States
| | - Walter J. Lukiw
- Alchem Biotech Research, Toronto, ON, Canada
- LSU Neuroscience Center, Louisiana State University Health Science Center, New Orleans, LA, United States
- Department of Ophthalmology, Louisiana State University Health Science Center, New Orleans, LA, United States
- Department Neurology, Louisiana State University Health Science Center, New Orleans, LA, United States
- *Correspondence: Walter J. Lukiw
| |
Collapse
|
111
|
Conditioned medium from amniotic fluid mesenchymal stem cells could modulate Alzheimer's disease-like changes in human neuroblastoma cell line SY-SY5Y in a paracrine manner. Tissue Cell 2022; 76:101808. [DOI: 10.1016/j.tice.2022.101808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 04/23/2022] [Accepted: 04/24/2022] [Indexed: 11/19/2022]
|
112
|
Ruiz-Gonzalez C, Cardona D, Rodriguez-Arrastia M, Ropero-Padilla C, Rueda-Ruzafa L, Carvajal F, Sanchez-Labraca N, Aparicio Mota A, Roman P. Effects of probiotics on cognitive and emotional functions in healthy older adults: Protocol for a double-blind randomized placebo-controlled crossover trial. Res Nurs Health 2022; 45:274-286. [PMID: 35080033 DOI: 10.1002/nur.22209] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 01/08/2022] [Accepted: 01/12/2022] [Indexed: 12/17/2022]
Abstract
Aging is a process that includes changes in cognitive and emotional functions, as well as changes in the diversity and integrity of gut microbiota. Probiotic treatments have recently been studied as a potential new therapeutic approach to alleviate a wide range of problems in other populations; however, clinical studies in older adults remain insufficient and limited. Thus, the aim of this project is to evaluate the efficacy of a multispecies probiotic formulation as a therapeutic strategy for attenuating the emotional and cognitive decline associated with aging in adults over the age of 55. This is a double-blind randomized placebo-controlled crossover trial involving at least 32 older adults and comparing two conditions: (a) probiotic, providing a multispecies probiotic for 10 weeks (Lactobacillus rhamnosus and Bifidobacterium lactis); and (b) placebo, receiving a harmless substance (potato starch). Despite the increasing use of probiotics for the treatment of cognitive and emotional problems, no study has yet focused on this group, to the best of our knowledge. Therapeutic strategies of the kind outlined in this protocol will help to shed light on the current state of knowledge about this topic, as well as promote health programs tailored to this population, which would encourage active aging and healthy lifestyles. Not only do we expect improvements in the emotional dimension in terms of anxiety, stress, depression, and sleep quality, we also expect improvements in the cognitive dimension in terms of attention, memory, and decreased impulsivity.
Collapse
Affiliation(s)
- Cristofer Ruiz-Gonzalez
- Department of Nursing Science, Physiotherapy and Medicine, Faculty of Health Sciences, University of Almeria, Almeria, Spain
| | - Diana Cardona
- Department of Nursing Science, Physiotherapy and Medicine, Faculty of Health Sciences, University of Almeria, Almeria, Spain.,Health Research Center CEINSA, University of Almeria, Almeria, Spain
| | - Miguel Rodriguez-Arrastia
- Pre-Department of Nursing, Faculty of Health Sciences, Jaume I University, Castello de la Plana, Spain.,Research Group CYS, Faculty of Health Sciences, Jaume I University, Castello de la Plana, Spain
| | - Carmen Ropero-Padilla
- Pre-Department of Nursing, Faculty of Health Sciences, Jaume I University, Castello de la Plana, Spain.,Research Group CYS, Faculty of Health Sciences, Jaume I University, Castello de la Plana, Spain
| | | | - Francisca Carvajal
- Health Research Center CEINSA, University of Almeria, Almeria, Spain.,Department of Psychology, Faculty of Psychology, University of Almeria, Almeria, Spain
| | - Nuria Sanchez-Labraca
- Department of Nursing Science, Physiotherapy and Medicine, Faculty of Health Sciences, University of Almeria, Almeria, Spain
| | - Adrian Aparicio Mota
- Fundación Pública Andaluza para la Investigación Biosanitaria de Andalucía Oriental (FIBAO), University Hospital Torrecárdenas, Almería, Spain
| | - Pablo Roman
- Department of Nursing Science, Physiotherapy and Medicine, Faculty of Health Sciences, University of Almeria, Almeria, Spain.,Health Research Center CEINSA, University of Almeria, Almeria, Spain.,Research Group CTS-451 Health Sciences, University of Almeria, Almeria, Spain
| |
Collapse
|
113
|
Nandwana V, Nandwana NK, Das Y, Saito M, Panda T, Das S, Almaguel F, Hosmane NS, Das BC. The Role of Microbiome in Brain Development and Neurodegenerative Diseases. Molecules 2022; 27:3402. [PMID: 35684340 PMCID: PMC9182002 DOI: 10.3390/molecules27113402] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/18/2022] [Accepted: 05/23/2022] [Indexed: 12/13/2022] Open
Abstract
Hundreds of billions of commensal microorganisms live in and on our bodies, most of which colonize the gut shortly after birth and stay there for the rest of our lives. In animal models, bidirectional communications between the central nervous system and gut microbiota (Gut-Brain Axis) have been extensively studied, and it is clear that changes in microbiota composition play a vital role in the pathogenesis of various neurodevelopmental and neurodegenerative disorders, such as Autism Spectrum Disorder, Alzheimer's disease, Parkinson's disease, Multiple Sclerosis, Amyotrophic Lateral Sclerosis, anxiety, stress, and so on. The makeup of the microbiome is impacted by a variety of factors, such as genetics, health status, method of delivery, environment, nutrition, and exercise, and the present understanding of the role of gut microbiota and its metabolites in the preservation of brain functioning and the development of the aforementioned neurological illnesses is summarized in this review article. Furthermore, we discuss current breakthroughs in the use of probiotics, prebiotics, and synbiotics to address neurological illnesses. Moreover, we also discussed the role of boron-based diet in memory, boron and microbiome relation, boron as anti-inflammatory agents, and boron in neurodegenerative diseases. In addition, in the coming years, boron reagents will play a significant role to improve dysbiosis and will open new areas for researchers.
Collapse
Affiliation(s)
- Varsha Nandwana
- Arnold and Marie Schwartz College of Pharmacy and Health Sciences, Long Island University, Brooklyn, NY 11201, USA; (V.N.); (N.K.N.); (T.P.); (S.D.)
| | - Nitesh K. Nandwana
- Arnold and Marie Schwartz College of Pharmacy and Health Sciences, Long Island University, Brooklyn, NY 11201, USA; (V.N.); (N.K.N.); (T.P.); (S.D.)
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Yogarupa Das
- Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA; (Y.D.); (M.S.)
| | - Mariko Saito
- Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA; (Y.D.); (M.S.)
| | - Tanisha Panda
- Arnold and Marie Schwartz College of Pharmacy and Health Sciences, Long Island University, Brooklyn, NY 11201, USA; (V.N.); (N.K.N.); (T.P.); (S.D.)
| | - Sasmita Das
- Arnold and Marie Schwartz College of Pharmacy and Health Sciences, Long Island University, Brooklyn, NY 11201, USA; (V.N.); (N.K.N.); (T.P.); (S.D.)
| | - Frankis Almaguel
- School of Medicine, Loma Linda University Health, Loma Linda, CA 92350, USA;
| | - Narayan S. Hosmane
- Department of Chemistry and Biochemistry, Northern Illinois University, DeKalb, IL 60115, USA;
| | - Bhaskar C. Das
- Arnold and Marie Schwartz College of Pharmacy and Health Sciences, Long Island University, Brooklyn, NY 11201, USA; (V.N.); (N.K.N.); (T.P.); (S.D.)
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
114
|
McGrattan A, Stewart CJ, Cassidy A, Woodside JV, McEvoy CT. Diet Patterns, the Gut Microbiome, and Alzheimer's Disease. J Alzheimers Dis 2022; 88:933-941. [PMID: 35634849 DOI: 10.3233/jad-220205] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Given the complex bidirectional communication system that exists between the gut microbiome and the brain, there is growing interest in the gut microbiome as a novel and potentially modifiable risk factor for Alzheimer's disease (AD). Gut dysbiosis has been implicated in the pathogenesis and progression of AD by initiating and prolonging neuroinflammatory processes. The metabolites of gut microbiota appear to be critical in the mechanism of the gut-brain axis. Gut microbiota metabolites, such as trimethylamine-n-oxide, lipopolysaccharide, and short chain fatty acids, are suggested to mediate systemic inflammation and intracerebral amyloidosis via endothelial dysfunction. Emerging data suggest that the fungal microbiota (mycobiome) may also influence AD pathology. Importantly, 60% of variation in the gut microbiome is attributable to diet, therefore modulating the gut microbiome through dietary means could be an effective approach to reduce AD risk. Given that people do not eat isolated nutrients and instead consume a diverse range of foods and combinations of nutrients that are likely to be interactive, studying the effects of whole diets provides the opportunity to account for the interactions between different nutrients. Thus, dietary patterns may be more predictive of real-life effect on gut microbiome and AD risk than foods or nutrients in isolation. Accumulating evidence from experimental and animal studies also show potential effects of gut microbiome on AD pathogenesis. However, data from human dietary interventions are lacking. Well-designed intervention studies are needed in diverse populations to determine the influence of diet on gut microbiome and inform the development of effective dietary strategies for prevention of AD.
Collapse
Affiliation(s)
- Andrea McGrattan
- School of Biomedical, Nutritional and Sport Sciences, Newcastle University, UK
| | | | - Aedín Cassidy
- Institute For Global Food Security, Queen's University Belfast, UK
| | - Jayne V Woodside
- Institute For Global Food Security, Queen's University Belfast, UK
| | - Claire T McEvoy
- Institute For Global Food Security, Queen's University Belfast, UK.,Global Brain Health Institute, University of California San Francisco, USA and Trinity College Dublin, Ireland
| |
Collapse
|
115
|
Giridharan VV, Barichello de Quevedo CE, Petronilho F. Microbiota-gut-brain axis in the Alzheimer's disease pathology - an overview. Neurosci Res 2022; 181:17-21. [DOI: 10.1016/j.neures.2022.05.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 05/02/2022] [Accepted: 05/10/2022] [Indexed: 12/12/2022]
|
116
|
Wiatrak B, Balon K, Jawień P, Bednarz D, Jęśkowiak I, Szeląg A. The Role of the Microbiota-Gut-Brain Axis in the Development of Alzheimer's Disease. Int J Mol Sci 2022; 23:ijms23094862. [PMID: 35563253 PMCID: PMC9104401 DOI: 10.3390/ijms23094862] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/16/2022] [Accepted: 04/21/2022] [Indexed: 02/06/2023] Open
Abstract
Along with the increase in life expectancy in the populations of developed and developing countries resulting from better access and improved health care, the number of patients with dementia, including Alzheimer’s disease (AD), is growing. The disease was first diagnosed and described at the beginning of the 20th century. However, to this day, there is no effective causal therapy, and symptomatic treatment often improves patients’ quality of life only for a short time. The current pharmacological therapies are based mainly on the oldest hypotheses of the disease—cholinergic (drugs affecting the cholinergic system are available), the hypothesis of amyloid-β aggregation (an anti-amyloid drug was conditionally approved by the FDA in 2020), and one drug is an N-methyl-D-aspartate receptor (NMDAR) antagonist (memantine). Hypotheses about AD pathogenesis focus on the nervous system and the brain. As research progresses, it has become known that AD can be caused by diseases that have been experienced over the course of a lifetime, which could also affect other organs. In this review, we focus on the potential association of AD with the digestive system, primarily the gut microbiota. The role of diet quality in preventing and alleviating Alzheimer’s disease is also discussed. The problem of neuroinflammation, which may be the result of microbiota disorders, is also described. An important aspect of the work is the chapter on the treatment strategies for changing the microbiota, potentially protecting against the disease and alleviating its course in the initial stages.
Collapse
Affiliation(s)
- Benita Wiatrak
- Department of Pharmacology, Wroclaw Medical University, Mikulicza-Radeckiego 2, 50-345 Wroclaw, Poland; (D.B.); (I.J.); (A.S.)
- Correspondence: (B.W.); (P.J.)
| | - Katarzyna Balon
- Laboratory of Genomics & Bioinformatics, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland;
| | - Paulina Jawień
- Department of Biostructure and Animal Physiology, Wroclaw University of Environmental and Life Sciences, Norwida 25/27, 50-375 Wroclaw, Poland
- Correspondence: (B.W.); (P.J.)
| | - Dominika Bednarz
- Department of Pharmacology, Wroclaw Medical University, Mikulicza-Radeckiego 2, 50-345 Wroclaw, Poland; (D.B.); (I.J.); (A.S.)
| | - Izabela Jęśkowiak
- Department of Pharmacology, Wroclaw Medical University, Mikulicza-Radeckiego 2, 50-345 Wroclaw, Poland; (D.B.); (I.J.); (A.S.)
| | - Adam Szeląg
- Department of Pharmacology, Wroclaw Medical University, Mikulicza-Radeckiego 2, 50-345 Wroclaw, Poland; (D.B.); (I.J.); (A.S.)
| |
Collapse
|
117
|
Histamine Causes Pyroptosis of Liver by Regulating Gut-Liver Axis in Mice. Int J Mol Sci 2022; 23:ijms23073710. [PMID: 35409071 PMCID: PMC8998596 DOI: 10.3390/ijms23073710] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 03/24/2022] [Accepted: 03/24/2022] [Indexed: 02/05/2023] Open
Abstract
Huangjiu usually caused rapid-drunkenness and components such as β-benzyl ethanol (β-be), isopentanol (Iso), histamine (His), and phenethylamine (PEA) have been reported linked with intoxication. However, the destructive effect of these components on gut microbiota and liver is unclear. In this study, we found oral treatment of these components, especially His, stimulated the level of oxidative stress and inflammatory cytokines in liver and serum of mice. The gut microbiota community was changed and the level of lipopolysaccharide (LPS) increased significantly. Additionally, cellular pyroptosis pathway has been assessed and correlation analysis revealed a possible relationship between gut microbiota and liver pyroptosis. We speculated oral His treatment caused the reprogramming of gut microbiota metabolism, and increased LPS modulated the gut-liver interaction, resulting in liver pyroptosis, which might cause health risks. This study provided a theoretical basis for the effect of Huangjiu, facilitating the development of therapeutic and preventive strategies for related inflammatory disorders.
Collapse
|
118
|
Kociszewska D, Vlajkovic SM. The Association of Inflammatory Gut Diseases with Neuroinflammatory and Auditory Disorders. Front Biosci (Elite Ed) 2022; 14:8. [PMID: 35730449 DOI: 10.31083/j.fbe1402008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/11/2022] [Accepted: 02/24/2022] [Indexed: 11/06/2022]
Abstract
Disorders such as inflammatory bowel disease (IBD) and celiac disease (CeD) result in intestinal hyperpermeability or 'leaky' gut. The increased permeability of the intestinal barrier allows microbial metabolites, toxins, and pathogens to infiltrate the bloodstream and extraintestinal tissues, causing systemic inflammation. Despite differences in aetiology and pathophysiology, IBD and CeD share several extraintestinal manifestations such as neuroinflammation, neurological and psychiatric manifestations, and sensorineural hearing loss (SNHL). This narrative review focuses on the association between intestinal hyperpermeability with the brain and inner ear diseases. We postulate that the microbial metabolites and pathogens released from the gut increase the permeability of natural barriers, such as the blood-brain barrier (BBB) and blood-labyrinth barrier (BLB). The barrier breakdown allows the spreading of inflammatory processes to the brain and inner ear, leading to disease.
Collapse
Affiliation(s)
- Dagmara Kociszewska
- Department of Physiology and The Eisdell Moore Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag, 1142 Auckland, New Zealand
| | - Srdjan M Vlajkovic
- Department of Physiology and The Eisdell Moore Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag, 1142 Auckland, New Zealand
| |
Collapse
|
119
|
Wu S, Wu Q, Wang J, Li Y, Chen B, Zhu Z, Huang R, Chen M, Huang A, Xie Y, Jiao C, Ding Y. Novel Selenium Peptides Obtained from Selenium-Enriched Cordyceps militaris Alleviate Neuroinflammation and Gut Microbiota Dysbacteriosis in LPS-Injured Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:3194-3206. [PMID: 35238567 DOI: 10.1021/acs.jafc.1c08393] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Increasing attention focuses on the relationship between neuroinflammation and Alzheimer's disease (AD). The reports on the microbiota-gut-brain axis reveal that the regulation by gut microbiota is an effective way to intervene in neuroinflammation-related AD. In this study, two novel selenium peptides (Se-Ps), VPRKL(Se)M (Se-P1) and RYNA(Se)MNDYT (Se-P2), with neuroprotection effects were obtained from Se-enriched Cordyceps militaris. Se-P1 and Se-P2 pre-protection led to a 30 and 33% increase in the PC-12 cell viability compared to the damage group, respectively. Moreover, Se-Ps exhibited a significant pre-protection against LPS-induced inflammatory and oxidative stress in the colon and brain by inhibiting the production of pro-inflammatory mediators (p < 0.05) and malondialdehyde, as well as promoting anti-inflammatory cytokine level and antioxidant enzyme activity (p < 0.05), which may alleviate the cognitive impairment in LPS-injured mice (p < 0.05). Se-Ps not only repaired the intestinal mucosa damage of LPS-injured mice but also had a positive effect on gut microbiota dysbacteriosis by increasing the abundance of Lactobacillus and Alistipes and decreasing the abundance of Akkermansia and Bacteroides. Collectively, the antioxidant, anti-inflammatory, and regulating properties on gut microflora of Se-Ps contribute to their neuroprotection, supporting that Se-Ps could be a promising dietary supplement in the prevention and/or treatment of AD.
Collapse
Affiliation(s)
- Shujian Wu
- Department of Food Science and Technology, Institute of Food Safety and Nutrition, College of Science & Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, P.R. China
- Institute of Microbiology, Guangdong Academy of Sciences, State Key Laboratory of Applied Microbiology Southern China, Guangdong, Guangdong Provincial Key Laboratory of Microbial Safety and Health, Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangzhou 510070, P.R. China
| | - Qingping Wu
- Institute of Microbiology, Guangdong Academy of Sciences, State Key Laboratory of Applied Microbiology Southern China, Guangdong, Guangdong Provincial Key Laboratory of Microbial Safety and Health, Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangzhou 510070, P.R. China
| | - Juan Wang
- College of Food Science, South China Agricultural University, Guangzhou 510070, P.R. China
| | - Yangfu Li
- Department of Food Science and Technology, Institute of Food Safety and Nutrition, College of Science & Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, P.R. China
- Institute of Microbiology, Guangdong Academy of Sciences, State Key Laboratory of Applied Microbiology Southern China, Guangdong, Guangdong Provincial Key Laboratory of Microbial Safety and Health, Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangzhou 510070, P.R. China
| | - Bo Chen
- Department of Food Science and Technology, Institute of Food Safety and Nutrition, College of Science & Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, P.R. China
- Institute of Microbiology, Guangdong Academy of Sciences, State Key Laboratory of Applied Microbiology Southern China, Guangdong, Guangdong Provincial Key Laboratory of Microbial Safety and Health, Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangzhou 510070, P.R. China
| | - Zhenjun Zhu
- Department of Food Science and Technology, Institute of Food Safety and Nutrition, College of Science & Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, P.R. China
| | - Rui Huang
- Department of Food Science and Technology, Institute of Food Safety and Nutrition, College of Science & Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, P.R. China
- Institute of Microbiology, Guangdong Academy of Sciences, State Key Laboratory of Applied Microbiology Southern China, Guangdong, Guangdong Provincial Key Laboratory of Microbial Safety and Health, Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangzhou 510070, P.R. China
| | - Mengfei Chen
- Department of Food Science and Technology, Institute of Food Safety and Nutrition, College of Science & Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, P.R. China
- Institute of Microbiology, Guangdong Academy of Sciences, State Key Laboratory of Applied Microbiology Southern China, Guangdong, Guangdong Provincial Key Laboratory of Microbial Safety and Health, Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangzhou 510070, P.R. China
| | - Aohuan Huang
- Department of Food Science and Technology, Institute of Food Safety and Nutrition, College of Science & Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, P.R. China
- Institute of Microbiology, Guangdong Academy of Sciences, State Key Laboratory of Applied Microbiology Southern China, Guangdong, Guangdong Provincial Key Laboratory of Microbial Safety and Health, Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangzhou 510070, P.R. China
| | - Yizhen Xie
- Guangdong Yuewei Edible Mushroom Technology Co., Ltd., Guangzhou 510700, P.R. China
| | - Chunwei Jiao
- Guangdong Yuewei Edible Mushroom Technology Co., Ltd., Guangzhou 510700, P.R. China
| | - Yu Ding
- Department of Food Science and Technology, Institute of Food Safety and Nutrition, College of Science & Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, P.R. China
| |
Collapse
|
120
|
SOCE-mediated NFAT1–NOX2–NLRP1 inflammasome involves in lipopolysaccharide-induced neuronal damage and Aβ generation. Mol Neurobiol 2022; 59:3183-3205. [DOI: 10.1007/s12035-021-02717-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 12/24/2021] [Indexed: 12/27/2022]
|
121
|
The Effects of Synbiotics Administration on Stress-Related Parameters in Thai Subjects-A Preliminary Study. Foods 2022; 11:foods11050759. [PMID: 35267392 PMCID: PMC8909555 DOI: 10.3390/foods11050759] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 03/01/2022] [Accepted: 03/03/2022] [Indexed: 12/04/2022] Open
Abstract
Urbanization influences our lifestyle, especially in fast-paced environments where we are more prone to stress. Stress management is considered advantageous in terms of longevity. The use of probiotics for psychological treatment has a small amount of diverse proven evidence to support this. However, studies on stress management in stressed subjects using synbiotics are still limited. The present study aimed to investigate the effects of synbiotics on stress in the Thai population. A total of 32 volunteers were enrolled and screened using a Thai Stress Test (TST) to determine their stress status. Participants were divided into the stressed and the non-stressed groups. Synbiotics preparation comprised a mixture of probiotics strains in a total concentration of 1 × 1010 CFU/day (5.0 × 109 CFU of Lactobacillus paracasei HII01 and 5.0 × 109 CFU of Bifidobacterium animalis subsp. lactis) and 10 g prebiotics (5 g galacto-oligosaccharides (GOS), and 5 g oligofructose (FOS)). All parameters were measured at baseline and after the 12th week of the study. In the stressed group, the administration of synbiotics significantly (p < 0.05) reduced the negative scale scores of TST, and tryptophan. In the non-stressed group, the synbiotics administration decreased tryptophan significantly (p < 0.05), whereas dehydroepiandrosterone sulfate (DHEA-S), tumor necrosis factor-α (TNF-α), 5-hydroxyindoleacetic acid (5-HIAA), and short-chain fatty acids (SCFAs), acetate and propionate were increased significantly (p < 0.05). In both groups, cortisol, and lipopolysaccharide (LPS) were reduced, whereas anti-inflammatory mediator interleukin-10 (IL-10) and immunoglobulin A (IgA) levels were increased. In conclusion, synbiotics administration attenuated the negative feelings via the negative scale scores of TST in stressed participants by modulating the HPA-axis, IL-10, IgA, and LPS. In comparison, synbiotics administration for participants without stress did not benefit stress status but showed remodeling SCFAs components, HPA-axis, and tryptophan catabolism.
Collapse
|
122
|
Murray ER, Kemp M, Nguyen TT. The Microbiota-Gut-Brain Axis in Alzheimer's Disease: A Review of Taxonomic Alterations and Potential Avenues for Interventions. Arch Clin Neuropsychol 2022; 37:595-607. [PMID: 35202456 PMCID: PMC9035085 DOI: 10.1093/arclin/acac008] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/22/2022] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVE The gut microbiome is a complex community of microorganisms that inhabit the gastrointestinal tract. The microbiota-gut-brain axis encompasses a bidirectional communication system that allows the gut to influence the brain via neural, endocrine, immune, and metabolic signaling. Differences in the gut microbiome have been associated with psychiatric and neurological disorders, including Alzheimer's Disease (ad). Understanding these ad-associated alterations may offer novel insight into the pathology and treatment of ad. METHOD We conducted a narrative review of clinical studies investigating the gut microbiome in ad, organizing the results by phyla to understand the biological contributions of the gut microbial community to ad pathology and clinical features. We also reviewed randomized clinical trials of interventions targeting the microbiome to ameliorate ad symptoms and biomarkers. RESULTS Alpha diversity is reduced in patients with ad. Within Firmicutes, taxa that produce beneficial metabolites are reduced in ad, including Clostridiaceae, Lachnospiraceae, Ruminococcus, and Eubacterium. Within Bacteroidetes, findings were mixed, with studies showing either reduced or increased abundance of Bacteroides in mild cognitive impairment or ad patients. Proteobacteria that produce toxins tend to be increased in ad patients, including Escherichia/Shigella. A Mediterranean-ketogenic dietary intervention significantly increased beneficial short-chain fatty acids and taxa that were inversely correlated with changes in ad pathological markers. Probiotic supplementation with Lactobacillus spp. and Bifidobacterium spp. improved cognitive function and reduced inflammatory and metabolic markers in patients with ad. CONCLUSIONS The gut microbiome may provide insight into ad pathology and be a novel target for intervention. Potential therapeutics include probiotics and dietary intervention.
Collapse
Affiliation(s)
- Emily R Murray
- Division of Biological Sciences, University of California at San Diego, La Jolla, CA, USA,Department of Psychiatry, University of California at San Diego, La Jolla, CA, USA
| | - Mylon Kemp
- Department of Psychiatry, University of California at San Diego, La Jolla, CA, USA
| | - Tanya T Nguyen
- Corresponding author at: Associate Professor of Psychiatry, University of California at San Diego, 9500 Gilman Drive #0664, La Jolla, CA 92093, USA. Tel.: +(858)-246-5347; fax: +(858)-543-5475.E-mail address: (T.T. Nguyen)
| |
Collapse
|
123
|
Sheng C, Yang K, He B, Du W, Cai Y, Han Y. Combination of gut microbiota and plasma amyloid-β as a potential index for identifying preclinical Alzheimer's disease: a cross-sectional analysis from the SILCODE study. Alzheimers Res Ther 2022; 14:35. [PMID: 35164860 PMCID: PMC8843023 DOI: 10.1186/s13195-022-00977-x] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 02/06/2022] [Indexed: 12/15/2022]
Abstract
Background Plasma amyloid-β (Aβ) may facilitate identification of individuals with brain amyloidosis. Gut microbial dysbiosis in Alzheimer’s disease (AD) is increasingly being recognized. However, knowledge about alterations of gut microbiota in preclinical AD, as well as whether the combination of plasma Aβ and gut microbiota could identify preclinical AD, remains largely unknown. Methods This study recruited 34 Aβ-negative cognitively normal (CN−) participants, 32 Aβ-positive cognitively normal (CN+) participants, and 22 patients with cognitive impairment (CI), including 11 patients with mild cognitive impairment (MCI) and 11 AD dementia patients. All participants underwent neuropsychological assessments and fecal microbiota analysis through 16S ribosomal RNA (rRNA) Illumina Miseq sequencing technique. Meso Scale Discovery (MSD) kits were used to quantify the plasma Aβ40, Aβ42, and Aβ42/Aβ40 in CN− and CN+ participants. Using Spearman’s correlation analysis, the associations of global standard uptake value rate (SUVR) with altered gut microbiota and plasma Aβ markers were separately evaluated. Furthermore, the discriminative power of the combination of gut microbiota and plasma Aβ markers for identifying CN+ individuals was investigated. Results Compared with the CN− group, the CN+ group showed significantly reduced plasma Aβ42 (p = 0.011) and Aβ42/Aβ40 (p = 0.003). The relative abundance of phylum Bacteroidetes was significantly enriched, whereas phylum Firmicutes and class Deltaproteobacteria were significantly decreased in CN+ individuals in comparison with that in CN− individuals. Particularly, the relative abundance of phylum Firmicutes and its corresponding SCFA-producing bacteria exhibited a progressive decline tendency from CN− to CN+ and CI. Besides, the global brain Aβ burden was negatively associated with the plasma Aβ42/Aβ40 (r = −0.298, p = 0.015), family Desulfovibrionaceae (r = −0.331, p = 0.007), genus Bilophila (r = −0.247, p = 0.046), and genus Faecalibacterium (r = −0.291, p = 0.018) for all CN participants. Finally, the combination of plasma Aβ markers, altered gut microbiota, and cognitive performance reached a relatively good discriminative power in identifying individuals with CN+ from CN− (AUC = 0.869, 95% CI 0.782 ~ 0.955). Conclusions This study provided the evidence that the gut microbial composition was altered in preclinical AD. The combination of plasma Aβ and gut microbiota may serve as a non-invasive, cost-effective diagnostic tool for early AD screening. Targeting the gut microbiota may be a novel therapeutic strategy for AD. Trial registration This study has been registered in ClinicalTrials.gov (NCT03370744, https://www.clinicaltrials.gov) in November 15, 2017. Supplementary Information The online version contains supplementary material available at 10.1186/s13195-022-00977-x.
Collapse
Affiliation(s)
- Can Sheng
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China
| | - Kun Yang
- Evidence-Based Medicine Center, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China.,Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, Beijing, 100069, China
| | - Beiqi He
- Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Haikou, 570228, China
| | - Wenying Du
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China
| | - Yanning Cai
- Department of Neurobiology, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China.,Key Laboratory for Neurodegenerative Diseases of the Ministry of Education, Beijing, 100053, China.,Department of Biobank, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China
| | - Ying Han
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China. .,Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Haikou, 570228, China. .,Center of Alzheimer's Disease, Beijing Institute for Brain Disorders, Beijing, 100053, China. .,National Clinical Research Center for Geriatric Diseases, Beijing, 100053, China.
| |
Collapse
|
124
|
Markoutsa E, Mayilsamy K, Gulick D, Mohapatra SS, Mohapatra S. Extracellular vesicles derived from inflammatory-educated stem cells reverse brain inflammation-implication of miRNAs. Mol Ther 2022; 30:816-830. [PMID: 34371179 PMCID: PMC8821927 DOI: 10.1016/j.ymthe.2021.08.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 07/04/2021] [Accepted: 07/29/2021] [Indexed: 02/04/2023] Open
Abstract
Inflammation plays a key role in the development of age-related diseases. In Alzheimer's disease, neuronal cell death is attributed to amyloidbeta oligomers that trigger microglial activation. Stem cells have shown promise as therapies for inflammatory diseases- because of their paracrine activity combined with their ability to respond to the inflammatory environment. However, the mechanisms underlying stem cell-promoted neurological recovery are poorly understood. To elucidate these mechanisms, we first primed stem cells with the secretome of lipopolysaccharide- or amyloidbeta-activated microglia. Then, we compared the immunomodulatory effects of extracellular vesicles (EVs) secreted from primed and non-primed stem cells. Our results demonstrate that EVs from primed cells are more effective in inhibiting microglia and astrocyte activation, amyloid deposition, demyelination, memory loss and motor and anxiety-like behavioral dysfunction, compared to EVs from non-primed cells. MicroRNA (miRNA) profiling revealed the upregulation of at least 19 miRNAs on primed-stem cell EVs. The miRNA targets were identified, and KEGG pathway analysis showed that the overexpressed miRNAs target key genes on the toll-like receptor-4 (TLR4) signaling pathway. Overall, our results demonstrate that priming mesenchymal stem cells (MSCs) with the secretome of activated microglia results in the release of miRNAs from EVs with enhanced immune regulatory potential able to fight neuroinflammation.
Collapse
Affiliation(s)
- Eleni Markoutsa
- James A. Haley VA Hospital, Tampa, FL 33612, USA,Division of Translational Medicine and Center for Research and Education in Nanobio-engineering, Department of Internal Medicine, University of South Florida Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA,College of Pharmacy Graduate Programs, University of South Florida, Tampa, FL, USA,Corresponding author: Eleni Markoutsa, Division of Translational Medicine and Center for Research and Education in Nanobio-engineering, Department of Internal Medicine, University of South Florida Mrsani College of Medicine, University of South Florida, Tampa, FL 33612, USA.
| | - Karthick Mayilsamy
- James A. Haley VA Hospital, Tampa, FL 33612, USA,Department of Molecular Medicine, University of South Florida Mrsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, USA
| | - Dannielle Gulick
- Department of Molecular Medicine, University of South Florida Mrsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, USA
| | - Shyam S. Mohapatra
- James A. Haley VA Hospital, Tampa, FL 33612, USA,Division of Translational Medicine and Center for Research and Education in Nanobio-engineering, Department of Internal Medicine, University of South Florida Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA,College of Pharmacy Graduate Programs, University of South Florida, Tampa, FL, USA
| | - Subhra Mohapatra
- James A. Haley VA Hospital, Tampa, FL 33612, USA,Department of Molecular Medicine, University of South Florida Mrsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, USA,Corresponding author: Subhra Mohapatra, Department of Molecular Medicine, University of South Florida Mrsani College of Medicine, University of South Florida, Tampa, FL 33612, USA.
| |
Collapse
|
125
|
Suhocki PV, Ronald JS, Diehl AME, Murdoch DM, Doraiswamy PM. Probing gut-brain links in Alzheimer's disease with rifaximin. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2022; 8:e12225. [PMID: 35128026 PMCID: PMC8804600 DOI: 10.1002/trc2.12225] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 11/05/2021] [Indexed: 12/12/2022]
Abstract
Gut-microbiome-inflammation interactions have been linked to neurodegeneration in Alzheimer's disease (AD) and other disorders. We hypothesized that treatment with rifaximin, a minimally absorbed gut-specific antibiotic, may modify the neurodegenerative process by changing gut flora and reducing neurotoxic microbial drivers of inflammation. In a pilot, open-label trial, we treated 10 subjects with mild to moderate probable AD dementia (Mini-Mental Status Examination (MMSE) = 17 ± 3) with rifaximin for 3 months. Treatment was associated with a significant reduction in serum neurofilament-light levels (P < .004) and a significant increase in fecal phylum Firmicutes microbiota. Serum phosphorylated tau (pTau)181 and glial fibrillary acidic protein (GFAP) levels were reduced (effect sizes of -0.41 and -0.48, respectively) but did not reach statistical significance. In addition, there was a nonsignificant downward trend in serum cytokine interleukin (IL)-6 and IL-13 levels. Cognition was unchanged. Increases in stool Erysipelatoclostridium were correlated significantly with reductions in serum pTau181 and serum GFAP. Insights from this pilot trial are being used to design a larger placebo-controlled clinical trial to determine if specific microbial flora/products underlie neurodegeneration, and whether rifaximin is clinically efficacious as a therapeutic.
Collapse
Affiliation(s)
| | | | | | | | - P. Murali Doraiswamy
- Duke University School of MedicineDurhamNorth CarolinaUSA
- Duke Institute for Brain SciencesDurhamNorth CarolinaUSA
| |
Collapse
|
126
|
Wan J, Zhou S, Mea HJ, Guo Y, Ku H, Urbina BM. Emerging Roles of Microfluidics in Brain Research: From Cerebral Fluids Manipulation to Brain-on-a-Chip and Neuroelectronic Devices Engineering. Chem Rev 2022; 122:7142-7181. [PMID: 35080375 DOI: 10.1021/acs.chemrev.1c00480] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Remarkable progress made in the past few decades in brain research enables the manipulation of neuronal activity in single neurons and neural circuits and thus allows the decipherment of relations between nervous systems and behavior. The discovery of glymphatic and lymphatic systems in the brain and the recently unveiled tight relations between the gastrointestinal (GI) tract and the central nervous system (CNS) further revolutionize our understanding of brain structures and functions. Fundamental questions about how neurons conduct two-way communications with the gut to establish the gut-brain axis (GBA) and interact with essential brain components such as glial cells and blood vessels to regulate cerebral blood flow (CBF) and cerebrospinal fluid (CSF) in health and disease, however, remain. Microfluidics with unparalleled advantages in the control of fluids at microscale has emerged recently as an effective approach to address these critical questions in brain research. The dynamics of cerebral fluids (i.e., blood and CSF) and novel in vitro brain-on-a-chip models and microfluidic-integrated multifunctional neuroelectronic devices, for example, have been investigated. This review starts with a critical discussion of the current understanding of several key topics in brain research such as neurovascular coupling (NVC), glymphatic pathway, and GBA and then interrogates a wide range of microfluidic-based approaches that have been developed or can be improved to advance our fundamental understanding of brain functions. Last, emerging technologies for structuring microfluidic devices and their implications and future directions in brain research are discussed.
Collapse
Affiliation(s)
- Jiandi Wan
- Department of Chemical Engineering, University of California, Davis, California 95616, United States
| | - Sitong Zhou
- Department of Chemical Engineering, University of California, Davis, California 95616, United States
| | - Hing Jii Mea
- Department of Chemical Engineering, University of California, Davis, California 95616, United States
| | - Yaojun Guo
- Department of Electrical and Computer Engineering, University of California, Davis, California 95616, United States
| | - Hansol Ku
- Department of Electrical and Computer Engineering, University of California, Davis, California 95616, United States
| | - Brianna M Urbina
- Biochemistry, Molecular, Cellular and Developmental Biology Program, University of California, Davis, California 95616, United States
| |
Collapse
|
127
|
Kramer P. Mitochondria-Microbiota Interaction in Neurodegeneration. Front Aging Neurosci 2022; 13:776936. [PMID: 35002678 PMCID: PMC8733591 DOI: 10.3389/fnagi.2021.776936] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 11/30/2021] [Indexed: 11/13/2022] Open
Abstract
Alzheimer’s and Parkinson’s are the two best-known neurodegenerative diseases. Each is associated with the excessive aggregation in the brain and elsewhere of its own characteristic amyloid proteins. Yet the two afflictions have much in common and often the same amyloids play a role in both. These amyloids need not be toxic and can help regulate bile secretion, synaptic plasticity, and immune defense. Moreover, when they do form toxic aggregates, amyloids typically harm not just patients but their pathogens too. A major port of entry for pathogens is the gut. Keeping the gut’s microbe community (microbiota) healthy and under control requires that our cells’ main energy producers (mitochondria) support the gut-blood barrier and immune system. As we age, these mitochondria eventually succumb to the corrosive byproducts they themselves release, our defenses break down, pathogens or their toxins break through, and the side effects of inflammation and amyloid aggregation become problematic. Although it gets most of the attention, local amyloid aggregation in the brain merely points to a bigger problem: the systemic breakdown of the entire human superorganism, exemplified by an interaction turning bad between mitochondria and microbiota.
Collapse
Affiliation(s)
- Peter Kramer
- Department of General Psychology, University of Padua, Padua, Italy
| |
Collapse
|
128
|
Ganz T, Fainstein N, Elad A, Lachish M, Goldfarb S, Einstein O, Ben-Hur T. Microbial pathogens induce neurodegeneration in Alzheimer's disease mice: protection by microglial regulation. J Neuroinflammation 2022; 19:5. [PMID: 34991645 PMCID: PMC8740456 DOI: 10.1186/s12974-021-02369-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 12/20/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Neurodegeneration is considered the consequence of misfolded proteins' deposition. Little is known about external environmental effects on the neurodegenerative process. Infectious agent-derived pathogen-associated molecular patterns (PAMPs) activate microglia, key players in neurodegenerative diseases. We hypothesized that systemic microbial pathogens may accelerate neurodegeneration in Alzheimer's disease (AD) and that microglia play a central role in this process. METHODS We examined the effect of an infectious environment and of microbial Toll-like receptor (TLR) agonists on cortical neuronal loss and on microglial phenotype in wild type versus 5xFAD transgenic mice, carrying mutated genes associated with familial AD. RESULTS We examined the effect of a naturally bred environment on the neurodegenerative process. Earlier and accelerated cortical neuron loss occurred in 5xFAD mice housed in a natural ("dirty") environment than in a specific-pathogen-free (SPF) environment, without increasing the burden of Amyloid deposits and microgliosis. Neuronal loss occurred in a microglia-rich cortical region but not in microglia-poor CA regions of the hippocampus. Environmental exposure had no effect on cortical neuron density in wild-type mice. To model the neurodegenerative process caused by the natural infectious environment, we injected systemically the bacterial endotoxin lipopolysaccharide (LPS), a TLR4 agonist PAMP. LPS caused cortical neuronal death in 5xFAD, but not wt mice. We used the selective retinoic acid receptor α agonist Am580 to regulate microglial activation. In primary microglia isolated from 5xFAD mice, Am580 markedly attenuated TLR agonists-induced iNOS expression, without canceling their basic immune response. Intracerebroventricular delivery of Am580 in 5xFAD mice reduced significantly the fraction of (neurotoxic) iNOS + microglia and increased the fraction of (neuroprotective) TREM2 + microglia. Furthermore, intracerebroventricular delivery of Am580 prevented neurodegeneration induced by microbial TLR agonists. CONCLUSIONS Exposure to systemic infections causes neurodegeneration in brain regions displaying amyloid pathology and high local microglia density. AD brains exhibit increased susceptibility to microbial PAMPs' neurotoxicity, which accelerates neuronal death. Microglial modulation protects the brain from microbial TLR agonist PAMP-induced neurodegeneration.
Collapse
Affiliation(s)
- Tal Ganz
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Nina Fainstein
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Amit Elad
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Marva Lachish
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Smadar Goldfarb
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Ofira Einstein
- Department of Physical Therapy, Faculty of Health Sciences, Ariel University, Ariel, Israel
| | - Tamir Ben-Hur
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel.
- The Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel.
| |
Collapse
|
129
|
The Role of Gut-Derived Lipopolysaccharides and the Intestinal Barrier in Fatty Liver Diseases. J Gastrointest Surg 2022; 26:671-683. [PMID: 34734369 PMCID: PMC8926958 DOI: 10.1007/s11605-021-05188-7] [Citation(s) in RCA: 101] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Accepted: 10/14/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Hepatosteatosis is the earliest stage in the pathogenesis of nonalcoholic fatty (NAFLD) and alcoholic liver disease (ALD). As NAFLD is affecting 10-24% of the general population and approximately 70% of obese patients, it carries a large economic burden and is becoming a major reason for liver transplantation worldwide. ALD is a major cause of morbidity and mortality causing 50% of liver cirrhosis and 10% of liver cancer related death. Increasing evidence has accumulated that gut-derived factors play a crucial role in the development and progression of chronic liver diseases. METHODS A selective literature search was conducted in Medline and PubMed, using the terms "nonalcoholic fatty liver disease," "alcoholic liver disease," "lipopolysaccharide," "gut barrier," and "microbiome." RESULTS Gut dysbiosis and gut barrier dysfunction both contribute to chronic liver disease by abnormal regulation of the gut-liver axis. Thereby, gut-derived lipopolysaccharides (LPS) are a key factor in inducing the inflammatory response of liver tissue. The review further underlines that endotoxemia is observed in both NAFLD and ALD patients. LPS plays an important role in conducting liver damage through the LPS-TLR4 signaling pathway. Treatments targeting the gut microbiome and the gut barrier such as fecal microbiota transplantation (FMT), probiotics, prebiotics, synbiotics, and intestinal alkaline phosphatase (IAP) represent potential treatment modalities for NAFLD and ALD. CONCLUSIONS The gut-liver axis plays an important role in the development of liver disease. Treatments targeting the gut microbiome and the gut barrier have shown beneficial effects in attenuating liver inflammation and need to be further investigated.
Collapse
|
130
|
Eo H, Lee S, Kim SH, Ju IG, Huh E, Lim J, Park S, Oh MS. Petasites japonicus leaf extract inhibits Alzheimer's-like pathology through suppression of neuroinflammation. Food Funct 2022; 13:10811-10822. [DOI: 10.1039/d2fo01989b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Neuroinflammation is a crucial step involved in development and progression of Alzheimer's disease. The current study found that Petasites japonicus leaf extract inhibits neuroinflammation induced by lipopolysaccharides and amyloid beta oligomers.
Collapse
Affiliation(s)
- Hyeyoon Eo
- Department of Biomedical and Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Seungmin Lee
- Department of Biomedical and Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Seong Hye Kim
- Department of Biomedical and Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - In Gyoung Ju
- Department of Oriental Pharmaceutical Science and Kyung Hee East-West Pharmaceutical Research Institute, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Eugene Huh
- Department of Oriental Pharmaceutical Science and Kyung Hee East-West Pharmaceutical Research Institute, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jeongin Lim
- NATUREBIO Co., Ltd., Seoul Biohub Industry-Academic Cooperation Center, Seoul 02447, Republic of Korea
| | - Sangsu Park
- NATUREBIO Co., Ltd., Seoul Biohub Industry-Academic Cooperation Center, Seoul 02447, Republic of Korea
| | - Myung Sook Oh
- Department of Biomedical and Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Oriental Pharmaceutical Science and Kyung Hee East-West Pharmaceutical Research Institute, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Integrated Drug Development and Natural Products, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
131
|
Shen P, Ji S, Li X, Yang Q, Xu B, Wong CKC, Wang L, Li L. LPS-Induced Systemic Inflammation Caused mPOA-FSH/LH Disturbance and Impaired Testicular Function. Front Endocrinol (Lausanne) 2022; 13:886085. [PMID: 35813649 PMCID: PMC9259990 DOI: 10.3389/fendo.2022.886085] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 05/03/2022] [Indexed: 11/26/2022] Open
Abstract
Male reproductive function is key to the continuation of species and is under sophisticated regulation, challenged by various stressors including inflammation. In the lipopolysaccharide (LPS) intraperitoneal injection-induced acute systemic inflammation, male fecundity was compromised with decreased testosterone level, damaged spermatogenesis, and downregulations of testicular gene expression levels involved in steroidogenesis regulation and blood-testis barrier. It is also noteworthy that the testis is more sensitive to acute stress caused by LPS-induced systemic inflammation. LPS treatment resulted in lower testicular gene expression levels of steroidogenic acute regulatory protein, cholesterol side-chain cleavage enzyme, and cytochrome P450 family 11 subfamily B member 1 after LPS treatment, while no such decrease was found in the adrenal gland. In parallel to the significant decreases in testicular intercellular adhesion molecule 1, tight junction protein 1, and gap junction alpha-1 protein gene expression with LPS treatment, no decrease was found in the epididymis. In the brain, LPS treatment caused higher medial preoptic area (mPOA) activation in the hypothalamus, which is accompanied by elevated blood follicle-stimulating hormone (FSH) and luteinizing hormone (LH) levels, suggesting a disturbed hypothalamic-pituitary-gonad axis function. Besides mPOA, brain c-fos mapping and quantitative analysis demonstrated a broad activation of brain nuclei by LPS, including the anterior cingulate cortex, lateral septum, paraventricular nucleus of the hypothalamus, basolateral amygdala, ventral tegmental area, lateral habenular nucleus, locus coeruleus, Barrington's nucleus, and the nucleus of the solitary tract, accompanied by abnormal animal behavior. Our data showed that LPS-induced inflammation caused not only local testicular damage but also a systemic disturbance at the brain-testis axis level.
Collapse
Affiliation(s)
- Peilei Shen
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Shuqin Ji
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xulin Li
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Qingning Yang
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Bingxian Xu
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, United States
| | - Chris Kong Chu Wong
- Croucher Institute for Environmental Sciences, Department of Biology, Hong Kong Baptist University Hong Kong, Hong Kong SAR, China
- *Correspondence: Chris Kong Chu Wong, ; Liping Wang, ; Lei Li,
| | - Liping Wang
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, Shenzhen, China
- CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen, China
- Shenzhen-Hong Kong Institute of Brain Science–Shenzhen Fundamental Research Institutions, Shenzhen, China
- *Correspondence: Chris Kong Chu Wong, ; Liping Wang, ; Lei Li,
| | - Lei Li
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, Shenzhen, China
- CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen, China
- Shenzhen-Hong Kong Institute of Brain Science–Shenzhen Fundamental Research Institutions, Shenzhen, China
- *Correspondence: Chris Kong Chu Wong, ; Liping Wang, ; Lei Li,
| |
Collapse
|
132
|
Page MJ, Kell DB, Pretorius E. The Role of Lipopolysaccharide-Induced Cell Signalling in Chronic Inflammation. CHRONIC STRESS (THOUSAND OAKS, CALIF.) 2022; 6:24705470221076390. [PMID: 35155966 PMCID: PMC8829728 DOI: 10.1177/24705470221076390] [Citation(s) in RCA: 121] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 01/11/2022] [Indexed: 12/20/2022]
Abstract
Lipopolysaccharide (LPS) is the main structural component of the outer membrane of most Gram-negative bacteria and has diverse immunostimulatory and procoagulant effects. Even though LPS is well described for its role in the pathology of sepsis, considerable evidence demonstrates that LPS-induced signalling and immune dysregulation are also relevant in the pathophysiology of many diseases, characteristically where endotoxaemia is less severe. These diseases are typically chronic and progressive in nature and span broad classifications, including neurodegenerative, metabolic, and cardiovascular diseases. This Review reappraises the mechanisms of LPS-induced signalling and emphasises the crucial contribution of LPS to the pathology of multiple chronic diseases, beyond conventional sepsis. This perspective asserts that new ways of approaching chronic diseases by targeting LPS-driven pathways may be of therapeutic benefit in a wide range of chronic inflammatory conditions.
Collapse
Affiliation(s)
| | - Douglas B Kell
- Stellenbosch University, Stellenbosch, South Africa.,Institute of Integrative Biology, University of Liverpool, Liverpool, UK.,The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Lyngby, Denmark
| | | |
Collapse
|
133
|
Sohrabi M, Sahu B, Kaur H, Hasler WA, Prakash A, Combs CK. Gastrointestinal Changes and Alzheimer's Disease. Curr Alzheimer Res 2022; 19:335-350. [PMID: 35718965 PMCID: PMC10497313 DOI: 10.2174/1567205019666220617121255] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/08/2022] [Accepted: 03/06/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND There is a well-described mechanism of communication between the brain and gastrointestinal system in which both organs influence the function of the other. This bi-directional communication suggests that disease in either organ may affect function in the other. OBJECTIVE To assess whether the evidence supports gastrointestinal system inflammatory or degenerative pathophysiology as a characteristic of Alzheimer's disease (AD). METHODS A review of both rodent and human studies implicating gastrointestinal changes in AD was performed. RESULTS Numerous studies indicate that AD changes are not unique to the brain but also occur at various levels of the gastrointestinal tract involving both immune and neuronal changes. In addition, it appears that numerous conditions and diseases affecting regions of the tract may communicate to the brain to influence disease. CONCLUSION Gastrointestinal changes represent an overlooked aspect of AD, representing a more system influence of this disease.
Collapse
Affiliation(s)
- Mona Sohrabi
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202-9037, USA
| | - Bijayani Sahu
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202-9037, USA
| | - Harpreet Kaur
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202-9037, USA
| | - Wendie A Hasler
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202-9037, USA
| | - Atish Prakash
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202-9037, USA
| | - Colin K Combs
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202-9037, USA
| |
Collapse
|
134
|
Pons V, Rivest S. Targeting Systemic Innate Immune Cells as a Therapeutic Avenue for Alzheimer Disease. Pharmacol Rev 2022; 74:1-17. [PMID: 34987086 DOI: 10.1124/pharmrev.121.000400] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 08/13/2021] [Indexed: 12/12/2022] Open
Abstract
Alzheimer disease (AD) is the first progressive neurodegenerative disease worldwide, and the disease is characterized by an accumulation of amyloid in the brain and neurovasculature that triggers cognitive decline and neuroinflammation. The innate immune system has a preponderant role in AD. The last decade, scientists focused their efforts on therapies aiming to modulate innate immunity. The latter is of great interest, since they participate to the inflammation and phagocytose the amyloid in the brain and blood vessels. We and others have developed pharmacological approaches to stimulate these cells using various ligands. These include toll-like receptor 4, macrophage colony stimulating factor, and more recently nucleotide-binding oligomerization domain-containing 2 receptors. This review will discuss the great potential to take advantage of the innate immune system to fight naturally against amyloid β accumulation and prevent its detrimental consequence on brain functions and its vascular system. SIGNIFICANCE STATEMENT: The focus on amyloid β removal from the perivascular space rather than targeting CNS plaque formation and clearance represents a new direction with a great potential. Small molecules able to act at the level of peripheral immunity would constitute a novel approach for tackling aberrant central nervous system biology, one of which we believe would have the potential of generating a lot of interest.
Collapse
Affiliation(s)
- Vincent Pons
- Neuroscience Laboratory, CHU de Québec Research Center and Department of Molecular Medicine, Faculty of Medicine, Laval University, 2705 Laurier Boul., Québec City, QC G1V 4G2, Canada
| | - Serge Rivest
- Neuroscience Laboratory, CHU de Québec Research Center and Department of Molecular Medicine, Faculty of Medicine, Laval University, 2705 Laurier Boul., Québec City, QC G1V 4G2, Canada
| |
Collapse
|
135
|
Zhan X, Hakoupian M, Jin LW, Sharp FR. Lipopolysaccharide, Identified Using an Antibody and by PAS Staining, Is Associated With Corpora amylacea and White Matter Injury in Alzheimer's Disease and Aging Brain. Front Aging Neurosci 2021; 13:705594. [PMID: 34899263 PMCID: PMC8652352 DOI: 10.3389/fnagi.2021.705594] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 09/06/2021] [Indexed: 12/16/2022] Open
Abstract
Corpora amylacea (CA) increase in number and size with aging. Their origins and functions remain unknown. Previously, we found that Alzheimer's disease (AD) brains have more CA in the periventricular white matter (PVWM) compared to aging controls. In addition, CA is associated with neurodegeneration as indicated by colocalization of degraded myelin basic protein (dMBP) with periodic acid-Schiff (PAS), a CA marker. We also found that bacterial lipopolysaccharide is present in aging brains, with more LPS in AD compared with controls. Periodic acid-Schiff staining is used to identify CA by virtue of their high polysaccharide content. Despite the growing knowledge of CA as a contributor to AD pathology, the molecules that contribute to the polysaccharides in CA are not known. Notably, lipopolysaccharides (LPS) are important cell-surface polysaccharides found in all Gram-negative bacteria. However, it is unknown whether PAS could detect LPS, whether the LPS found in aging brains contribute to the polysaccharide found in CA, and whether LPS associate with myelin injury. In this study, we found that aging brains had a myelin deficit zone (MDZ) adjacent to the ventricles in PVWM. The MDZ contained vesicles, most of which were CA. LPS and dMBP levels were higher in AD than in control brains. LPS was colocalized with dMBP in the vesicles/CA, linking white matter injury with a bacterial pro-inflammatory molecule. The vesicles also contained oxidized fibers, C-reactive protein, NG2, and GALC, markers of oligodendrocyte precursor cells (OPCs) and oligodendrocyte cells (OLs), respectively. The vesicles/CA were surrounded by dense astrocyte processes in control and AD brains. LPS was co-localized with CA by double staining of PAS with LPS in aging brains. The relationship of LPS with PAS staining was confirmed by PAS staining of purified LPS on nitrocellulose membranes. These findings reveal that LPS is one of the polysaccharides found in CA which can be stained with PAS. In addition, vesicles/CA are associated with oxidized and damaged myelin. The LPS in these vesicles/CA may have contributed to this oxidative myelin damage and may have contributed to oxidative stress to OPCs and OLs which could impair the ability to repair damaged myelin in AD and control brains.
Collapse
Affiliation(s)
- Xinhua Zhan
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Marisa Hakoupian
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Lee-Way Jin
- Department of Pathology, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Frank R Sharp
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA, United States
| |
Collapse
|
136
|
Kim HS, Kim S, Shin SJ, Park YH, Nam Y, Kim CW, Lee KW, Kim SM, Jung ID, Yang HD, Park YM, Moon M. Gram-negative bacteria and their lipopolysaccharides in Alzheimer's disease: pathologic roles and therapeutic implications. Transl Neurodegener 2021; 10:49. [PMID: 34876226 PMCID: PMC8650380 DOI: 10.1186/s40035-021-00273-y] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 11/09/2021] [Indexed: 02/07/2023] Open
Abstract
Alzheimer's disease (AD) is the most serious age-related neurodegenerative disease and causes destructive and irreversible cognitive decline. Failures in the development of therapeutics targeting amyloid-β (Aβ) and tau, principal proteins inducing pathology in AD, suggest a paradigm shift towards the development of new therapeutic targets. The gram-negative bacteria and lipopolysaccharides (LPS) are attractive new targets for AD treatment. Surprisingly, an altered distribution of gram-negative bacteria and their LPS has been reported in AD patients. Moreover, gram-negative bacteria and their LPS have been shown to affect a variety of AD-related pathologies, such as Aβ homeostasis, tau pathology, neuroinflammation, and neurodegeneration. Moreover, therapeutic approaches targeting gram-negative bacteria or gram-negative bacterial molecules have significantly alleviated AD-related pathology and cognitive dysfunction. Despite multiple evidence showing that the gram-negative bacteria and their LPS play a crucial role in AD pathogenesis, the pathogenic mechanisms of gram-negative bacteria and their LPS have not been clarified. Here, we summarize the roles and pathomechanisms of gram-negative bacteria and LPS in AD. Furthermore, we discuss the possibility of using gram-negative bacteria and gram-negative bacterial molecules as novel therapeutic targets and new pathological characteristics for AD.
Collapse
Affiliation(s)
- Hyeon Soo Kim
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, 35365, Republic of Korea
| | - Sujin Kim
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, 35365, Republic of Korea
- Research Institute for Dementia Science, Konyang University, Daejeon, 35365, Republic of Korea
| | - Soo Jung Shin
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, 35365, Republic of Korea
| | - Yong Ho Park
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, 35365, Republic of Korea
| | - Yunkwon Nam
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, 35365, Republic of Korea
| | - Chae Won Kim
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, 35365, Republic of Korea
| | - Kang Won Lee
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, 35365, Republic of Korea
| | - Sung-Min Kim
- Dandi Bioscience Inc, 6th Floor of Real Company Building, 66, Achasan-ro, Sungdong-gu, Seoul, Republic of Korea
| | - In Duk Jung
- Dandi Bioscience Inc, 6th Floor of Real Company Building, 66, Achasan-ro, Sungdong-gu, Seoul, Republic of Korea
| | - Hyun Duk Yang
- Harvard Neurology Clinic, 294 Gwanggyojungang-ro, Suji-gu, Yongin, 16943, Republic of Korea.
| | - Yeong-Min Park
- Dandi Bioscience Inc, 6th Floor of Real Company Building, 66, Achasan-ro, Sungdong-gu, Seoul, Republic of Korea.
- Department of Immunology, School of Medicine, Konkuk University, 268, Chungwondaero, Chungju-si, Chungcheongbuk-do, Republic of Korea.
| | - Minho Moon
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, 35365, Republic of Korea.
- Research Institute for Dementia Science, Konyang University, Daejeon, 35365, Republic of Korea.
| |
Collapse
|
137
|
Laudanski K, Hajj J, Restrepo M, Siddiq K, Okeke T, Rader DJ. Dynamic Changes in Central and Peripheral Neuro-Injury vs. Neuroprotective Serum Markers in COVID-19 Are Modulated by Different Types of Anti-Viral Treatments but Do Not Affect the Incidence of Late and Early Strokes. Biomedicines 2021; 9:1791. [PMID: 34944606 PMCID: PMC8698659 DOI: 10.3390/biomedicines9121791] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 11/15/2021] [Accepted: 11/19/2021] [Indexed: 01/07/2023] Open
Abstract
The balance between neurodegeneration, neuroinflammation, neuroprotection, and COVID-19-directed therapy may underly the heterogeneity of SARS-CoV-2's neurological outcomes. A total of 105 patients hospitalized with a diagnosis of COVID-19 had serum collected over a 6 month period to assess neuroinflammatory (MIF, CCL23, MCP-1), neuro-injury (NFL, NCAM-1), neurodegenerative (KLK6, τ, phospho τ, amyloids, TDP43, YKL40), and neuroprotective (clusterin, fetuin, TREM-2) proteins. These were compared to markers of nonspecific inflammatory responses (IL-6, D-dimer, CRP) and of the overall viral burden (spike protein). Data regarding treatment (steroids, convalescent plasma, remdasavir), pre-existing conditions, and incidences of strokes were collected. Amyloid β42, TDP43, NF-L, and KLK6 serum levels declined 2-3 days post-admission, yet recovered to admission baseline levels by 7 days. YKL-40 and NCAM-1 levels remained elevated over time, with clusters of differential responses identified among TREM-2, TDP43, and YKL40. Fetuin was elevated after the onset of COVID-19 while TREM-2 initially declined before significantly increasing over time. MIF serum level was increased 3-7 days after admission. Ferritin correlated with TDP-43 and KLK6. No treatment with remdesivir coincided with elevations in Amyloid-β40. A lack of convalescent plasma resulted in increased NCAM-1 and total tau, and steroidal treatments did not significantly affect any markers. A total of 11 incidences of stroke were registered up to six months after initial admission for COVID-19. Elevated D-dimer, platelet counts, IL-6, and leukopenia were observed. Variable MIF serum levels differentiated patients with CVA from those who did not have a stroke during the acute phase of COVID-19. This study demonstrated concomitant and opposite changes in neurodegenerative and neuroprotective markers persisting well into recovery.
Collapse
Affiliation(s)
- Krzysztof Laudanski
- The Department of Anesthesiology and Critical Care Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Neurology, University of Pennsylvania, Philadelphia, PA 19104, USA
- The Leonard Davis Institute of Health Economics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jihane Hajj
- School of Nursing, Widener University, Philadelphia, PA 19013, USA;
| | - Mariana Restrepo
- College of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Kumal Siddiq
- College of Arts and Sciences, Drexel University, Philadelphia, PA 19104, USA;
| | - Tony Okeke
- School of Biomedical Engineering, Drexel University, Philadelphia, PA 19104, USA;
| | - Daniel J. Rader
- Department of Genetics, University of Pennsylvania, Philadelphia, PA 19104, USA;
| |
Collapse
|
138
|
Frausto DM, Forsyth CB, Keshavarzian A, Voigt RM. Dietary Regulation of Gut-Brain Axis in Alzheimer's Disease: Importance of Microbiota Metabolites. Front Neurosci 2021; 15:736814. [PMID: 34867153 PMCID: PMC8639879 DOI: 10.3389/fnins.2021.736814] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 10/18/2021] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease that impacts 45 million people worldwide and is ranked as the 6th top cause of death among all adults by the Centers for Disease Control and Prevention. While genetics is an important risk factor for the development of AD, environment and lifestyle are also contributing risk factors. One such environmental factor is diet, which has emerged as a key influencer of AD development/progression as well as cognition. Diets containing large quantities of saturated/trans-fats, refined carbohydrates, limited intake of fiber, and alcohol are associated with cognitive dysfunction while conversely diets low in saturated/trans-fats (i.e., bad fats), high mono/polyunsaturated fats (i.e., good fats), high in fiber and polyphenols are associated with better cognitive function and memory in both humans and animal models. Mechanistically, this could be the direct consequence of dietary components (lipids, vitamins, polyphenols) on the brain, but other mechanisms are also likely to be important. Diet is considered to be the single greatest factor influencing the intestinal microbiome. Diet robustly influences the types and function of micro-organisms (called microbiota) that reside in the gastrointestinal tract. Availability of different types of nutrients (from the diet) will favor or disfavor the abundance and function of certain groups of microbiota. Microbiota are highly metabolically active and produce many metabolites and other factors that can affect the brain including cognition and the development and clinical progression of AD. This review summarizes data to support a model in which microbiota metabolites influence brain function and AD.
Collapse
Affiliation(s)
- Dulce M. Frausto
- Rush Medical College, Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
| | - Christopher B. Forsyth
- Rush Medical College, Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
- Department of Medicine, Rush University Medical Center, Chicago, IL, United States
| | - Ali Keshavarzian
- Rush Medical College, Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
- Department of Medicine, Rush University Medical Center, Chicago, IL, United States
- Department of Physiology, Rush University Medical Center, Chicago, IL, United States
| | - Robin M. Voigt
- Rush Medical College, Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
- Department of Medicine, Rush University Medical Center, Chicago, IL, United States
| |
Collapse
|
139
|
Kowalski K, Mulak A. Small intestinal bacterial overgrowth in Alzheimer's disease. J Neural Transm (Vienna) 2021; 129:75-83. [PMID: 34797427 PMCID: PMC8738624 DOI: 10.1007/s00702-021-02440-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 11/04/2021] [Indexed: 11/21/2022]
Abstract
The results of animal studies and clinical data support the gut microbiota contribution to the pathogenesis of Alzheimer’s disease (AD). The aim of this pilot study was to evaluate the prevalence of small intestinal bacterial overgrowth (SIBO) and fecal markers of intestinal inflammation and permeability in AD patients. The study was conducted in 45 AD patients and 27 controls. Data on comorbidities, pharmacotherapy, and gastrointestinal symptoms were acquired from medical records and a questionnaire. SIBO was evaluated using lactulose hydrogen breath test. Fecal calprotectin and zonulin levels were assessed by ELISA assays. The positive result of SIBO breath test was found in 49% of the AD patients and 22% of the controls (p = 0.025). The comparative analysis between SIBO-positive and SIBO-negative AD patients with respect to the degree of cognitive impairment, comorbidities and used medications did not reveal any statistically significant difference, except for less common heartburn in SIBO-positive AD patients than in SIBO-negative ones (9 vs 35%, p = 0.038). The median fecal calprotectin and zonulin levels in the AD group compared to the control group amounted to 43.1 vs 64.2 µg/g (p = 0.846) and 73.5 vs 49.0 ng/ml (p = 0.177), respectively. In the AD patients there was no association between the presence of SIBO and fecal calprotectin level. Patients with AD are characterized by higher prevalence of SIBO not associated with increased fecal calprotectin level that may be related to anti-inflammatory effect of cholinergic drugs used in the treatment of AD.
Collapse
Affiliation(s)
- Karol Kowalski
- Department of Gastroenterology and Hepatology, Wroclaw Medical University, Borowska 213, 50-556, Wrocław, Poland
| | - Agata Mulak
- Department of Gastroenterology and Hepatology, Wroclaw Medical University, Borowska 213, 50-556, Wrocław, Poland.
| |
Collapse
|
140
|
Spirulina platensis alleviates high fat diet-induced cognitive impairment in mice via the gut-brain axis. J Funct Foods 2021. [DOI: 10.1016/j.jff.2021.104706] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
141
|
Lee DY, Shin YJ, Kim JK, Jang HM, Joo MK, Kim DH. Alleviation of cognitive impairment by gut microbiota lipopolysaccharide production-suppressing Lactobacillus plantarum and Bifidobacterium longum in mice. Food Funct 2021; 12:10750-10763. [PMID: 34608923 DOI: 10.1039/d1fo02167b] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Bacterial lipopolysaccharide (LPS) is a risk factor for the outbreak of Alzheimer's disease. Therefore, we isolated Lactobacillus plantarum NK151 and Bifidobacterium longum NK173 from a human fecal bacteria collection, which inhibited Escherichia coli LPS production, and examined their effects on the Escherichia coli K1- or LPS-induced cognitive impairment in mice. Oral gavage of NK151, NK173, or their (4 : 1) mixture (NKm) significantly alleviated Escherichia coli K1-induced cognitive impairment-like behaviors in the Y-maze and novel object recognition tasks. Their treatments decreased IL-1β, IL-6, and TNF-α expression and NF-κB+/Iba1+ and LPS+/Iba1+ cell populations in the hippocampus, while the brain-derived neurotrophic factor (BDNF)+/neuronal nuclei (NeuN)+ cell population and BDNF to proBNDF expression increased. They suppressed LPS-induced cognition impairment-like behaviors and neuroinflammation marker levels in the hippocampus. Treatment with them reduced Escherichia coli K1- or LPS-induced LPS and apolipoprotein E levels in the blood and inflammatory marker levels in the colon. Furthermore, treatment with them modulated fecal Proteobacteria, Bacteroidetes, and Verrucomicrobia populations. Of these gut bacteria, Bacteroidaceae, Odoribacteraceae, Lactobacillaceae, Bifidobacteriaceae, Rikenellaceae, Helicobacteraceae, and Deferribacteraceae are correlated with cognitive function and blood and fecal LPS levels. These findings suggest that NK151 and NK173 may alleviate cognitive impairment with colitis by upregulating NF-κB-mediated BDNF expression through the suppression of fecal and blood bacterial LPS levels.
Collapse
Affiliation(s)
- Dong-Yun Lee
- Neurobiota Research Center, College of Pharmacy, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea.
| | - Yoon-Jung Shin
- Neurobiota Research Center, College of Pharmacy, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea.
| | - Jeon-Kyung Kim
- Neurobiota Research Center, College of Pharmacy, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea.
| | - Hyo-Min Jang
- Neurobiota Research Center, College of Pharmacy, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea.
| | - Min-Kyung Joo
- Neurobiota Research Center, College of Pharmacy, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea.
| | - Dong-Hyun Kim
- Neurobiota Research Center, College of Pharmacy, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea.
| |
Collapse
|
142
|
Ullah R, Park TJ, Huang X, Kim MO. Abnormal amyloid beta metabolism in systemic abnormalities and Alzheimer's pathology: Insights and therapeutic approaches from periphery. Ageing Res Rev 2021; 71:101451. [PMID: 34450351 DOI: 10.1016/j.arr.2021.101451] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 08/12/2021] [Accepted: 08/19/2021] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease (AD) is an age-associated, multifactorial neurodegenerative disorder that is incurable. Despite recent success in treatments that partially improve symptomatic relief, they have failed in most clinical trials. Re-holding AD for accurate diagnosis and treatment is widely known as a challenging task. Lack of knowledge of basic molecular pathogenesis might be a possible reason for ineffective AD treatment. Historically, a majority of therapy-based studies have investigated the role of amyloid-β (Aβ peptide) in the central nervous system (CNS), whereas less is known about Aβ peptide in the periphery in AD. In this review, we provide a comprehensive summary of the current understanding of Aβ peptide metabolism (anabolism and catabolism) in the brain and periphery. We show that the abnormal metabolism of Aβ peptide is significantly linked with central-brain and peripheral abnormalities; the interaction between peripheral Aβ peptide metabolism and peripheral abnormalities affects central-brain Aβ peptide metabolism, suggesting the existence of significant communication between these two pathways of Aβ peptide metabolism. This close interaction between the central brain and periphery in abnormal Aβ peptide metabolism plays a key role in the development and progression of AD. In conclusion, we need to obtain a full understanding of the dynamic roles of Aβ peptide at the molecular level in both the brain and periphery in relation to the pathology of AD. This will not only provide new information regarding the complex disease pathology, but also offer potential new clues to improve therapeutic strategies and diagnostic biomarkers for the successful treatment of AD.
Collapse
|
143
|
Khodabakhsh P, Bazrgar M, Dargahi L, Mohagheghi F, Asgari Taei A, Parvardeh S, Ahmadiani A. Does Alzheimer's disease stem in the gastrointestinal system? Life Sci 2021; 287:120088. [PMID: 34715145 DOI: 10.1016/j.lfs.2021.120088] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 10/13/2021] [Accepted: 10/21/2021] [Indexed: 02/07/2023]
Abstract
Over the last decades, our knowledge of the key pathogenic mechanisms of Alzheimer's disease (AD) has dramatically improved. Regarding the limitation of current therapeutic strategies for the treatment of multifactorial diseases, such as AD, to be translated into the clinic, there is a growing trend in research to identify risk factors associated with the onset and progression of AD. Here, we review the current literature with a focus on the relationship between gastrointestinal (GI)/liver diseases during the lifespan and the incidence of AD, and discuss the possible mechanisms underlying the link between the diseases. We also aim to review studies evaluating the possible link between the chronic use of the most common GI medications and the future risk of AD development.
Collapse
Affiliation(s)
- Pariya Khodabakhsh
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Bazrgar
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Leila Dargahi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Mohagheghi
- Institute of Experimental Hematology, Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany
| | - Afsaneh Asgari Taei
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Siavash Parvardeh
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Abolhassan Ahmadiani
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
144
|
Zhao Y, Arceneaux L, Culicchia F, Lukiw WJ. Neurofilament Light (NF-L) Chain Protein from a Highly Polymerized Structural Component of the Neuronal Cytoskeleton to a Neurodegenerative Disease Biomarker in the Periphery. HSOA JOURNAL OF ALZHEIMER'S & NEURODEGENERATIVE DISEASES 2021; 7:056. [PMID: 34881359 PMCID: PMC8651065 DOI: 10.24966/and-9608/100056] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neurofilaments (NFs) are critical scaffolding components of the axoskeleton of healthy neurons interacting directly with multiple synaptic-phosphoproteins to support and coordinate neuronal cell shape, cytoarchitecture, synaptogenesis and neurotransmission. While neuronal presynaptic proteins such as synapsin-2 (SYN II) degrade rapidly via the ubiquitin-proteasome pathway, a considerably more stable neurofilament light (NF-L) chain protein turns over much more slowly, and in several neurological diseases is accompanied by a pathological shift from an intracellular neuronal cytoplasmic location into various biofluid compartments. NF-L has been found to be significantly elevated in peripheral biofluids in multiple neurodegenerative disorders, however it is not as widely appreciated that NF-L expression within neurons undergoing inflammatory neurodegeneration exhibit a significant down-regulation in these neuron-specific intermediate-filament components. Down-regulated NF-L in neurons correlates well with the observed axonal and neuronal atrophy, neurite deterioration and synaptic disorganization in tissues affected by Alzheimer's disease (AD) and other progressive, age-related neurological diseases. This Review paper: (i) will briefly assess the remarkably high number of neurological disorders that exhibit NF-L depolymerization, liberation from neuron-specific compartments, mobilization and enrichment into pathological biofluids; (ii) will evaluate how NF-L exhibits compartmentalization effects in age-related neurological disorders; (iii) will review how the shift of NF-L compartmentalization from within the neuronal cytoskeleton into peripheral biofluids may be a diagnostic biomarker for neuronal-decline in all cause dementia most useful in distinguishing between closely related neurological disorders; and (iv) will review emerging evidence that deficits in plasma membrane barrier integrity, pathological transport and/or vesicle-mediated trafficking dysfunction of NF-L may contribute to neuronal decline, with specific reference to AD wherever possible.
Collapse
Affiliation(s)
- Yuhai Zhao
- LSU Neuroscience Center, Louisiana State University Health Science Center, New Orleans LA 70112, USA
- Department of Cell Biology and Anatomy, LSU Health Science Center, New Orleans LA 70112, USA
| | - Lisa Arceneaux
- LSU Neuroscience Center, Louisiana State University Health Science Center, New Orleans LA 70112, USA
| | - Frank Culicchia
- LSU Neuroscience Center, Louisiana State University Health Science Center, New Orleans LA 70112, USA
- Department of Neurosurgery, Louisiana State University Health Science Center, New Orleans LA 70112, USA
| | - Walter J Lukiw
- LSU Neuroscience Center, Louisiana State University Health Science Center, New Orleans LA 70112, USA
- Department of Ophthalmology, Louisiana State University Health Science Center, New Orleans LA 7011, USA
- Department of Neurology, Louisiana State University Health Science Center, New Orleans LA 70112, USA
| |
Collapse
|
145
|
Motolani A, Martin M, Sun M, Lu T. The Structure and Functions of PRMT5 in Human Diseases. Life (Basel) 2021; 11:life11101074. [PMID: 34685445 PMCID: PMC8539453 DOI: 10.3390/life11101074] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/06/2021] [Accepted: 10/07/2021] [Indexed: 12/17/2022] Open
Abstract
Since the discovery of protein arginine methyltransferase 5 (PRMT5) and the resolution of its structure, an increasing number of papers have investigated and delineated the structural and functional role of PRMT5 in diseased conditions. PRMT5 is a type II arginine methyltransferase that catalyzes symmetric dimethylation marks on histones and non-histone proteins. From gene regulation to human development, PRMT5 is involved in many vital biological functions in humans. The role of PRMT5 in various cancers is particularly well-documented, and investigations into the development of better PRMT5 inhibitors to promote tumor regression are ongoing. Notably, emerging studies have demonstrated the pathological contribution of PRMT5 in the progression of inflammatory diseases, such as diabetes, cardiovascular diseases, and neurodegenerative disorders. However, more research in this direction is needed. Herein, we critically review the position of PRMT5 in current literature, including its structure, mechanism of action, regulation, physiological and pathological relevance, and therapeutic strategies.
Collapse
Affiliation(s)
- Aishat Motolani
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.M.); (M.M.); (M.S.)
| | - Matthew Martin
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.M.); (M.M.); (M.S.)
| | - Mengyao Sun
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.M.); (M.M.); (M.S.)
| | - Tao Lu
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.M.); (M.M.); (M.S.)
- Department of Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Correspondence: ; Tel.: +1-317-278-0520
| |
Collapse
|
146
|
Nie JJ, Pian YY, Hu JH, Fan GQ, Zeng LT, Ouyang QG, Gao ZX, Liu Z, Wang CC, Liu Q, Cai JP. Increased systemic RNA oxidative damage and diagnostic value of RNA oxidative metabolites during Shigella flexneri-induced intestinal infection. World J Gastroenterol 2021; 27:6248-6261. [PMID: 34712030 PMCID: PMC8515791 DOI: 10.3748/wjg.v27.i37.6248] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/29/2021] [Accepted: 08/09/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Shigella flexneri (S. flexneri) is a major pathogen causing acute intestinal infection, but the systematic oxidative damage incurred during the course of infection has not been investigated.
AIM To investigate the incurred systemic RNA oxidative damage and the diagnostic value of RNA oxidative metabolites during S. flexneri-induced intestinal infection.
METHODS In this study, a Sprague-Dawley rat model of acute intestinal infection was established by oral gavage with S. flexneri strains. The changes in white blood cells (WBCs) and cytokine levels in blood and the inflammatory response in the colon were investigated. We also detected the RNA and DNA oxidation in urine and tissues.
RESULTS S. flexneri infection induced an increase in WBCs, C-reactive protein, interleukin (IL)-6, IL-10, IL-1β, IL-4, IL-17a, IL-10, and tumor necrosis factor α (TNF-α) in blood. Of note, a significant increase in urinary 8-oxo-7,8-dihydroguanosine (8-oxo-Gsn), an important marker of total RNA oxidation, was detected after intestinal infection (P = 0.03). The urinary 8-oxo-Gsn level returned to the baseline level after recovery from infection. In addition, the results of a correlation analysis showed that urinary 8-oxo-Gsn was positively correlated with the WBC count and the cytokines IL-6, TNF-α, IL-10, IL-1β, and IL-17α. Further detection of the oxidation in different tissues showed that S. flexneri infection induced RNA oxidative damage in the colon, ileum, liver, spleen, and brain.
CONCLUSION Acute infection induced by S. flexneri causes increased RNA oxidative damage in various tissues (liver, spleen, and brain) and an increase of 8-oxo-Gsn, a urinary metabolite. Urinary 8-oxo-Gsn may be useful as a biomarker for evaluating the severity and prognosis of infection.
Collapse
Affiliation(s)
- Jing-Jing Nie
- Department of Microbiology, National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Ya-Ya Pian
- Department of Microbiology, National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Ji-Hong Hu
- Department of Microbiology, National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Guo-Qing Fan
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Lv-Tao Zeng
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Qiu-Geng Ouyang
- Department of Pharmacy, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
| | - Zhen-Xiang Gao
- Department of Microbiology, National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Zhen Liu
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Chen-Chen Wang
- Department of Pharmacy, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
| | - Qian Liu
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Jian-Ping Cai
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, China
| |
Collapse
|
147
|
Mulvey CM, Breckels LM, Crook OM, Sanders DJ, Ribeiro ALR, Geladaki A, Christoforou A, Britovšek NK, Hurrell T, Deery MJ, Gatto L, Smith AM, Lilley KS. Spatiotemporal proteomic profiling of the pro-inflammatory response to lipopolysaccharide in the THP-1 human leukaemia cell line. Nat Commun 2021; 12:5773. [PMID: 34599159 PMCID: PMC8486773 DOI: 10.1038/s41467-021-26000-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 09/07/2021] [Indexed: 02/07/2023] Open
Abstract
Protein localisation and translocation between intracellular compartments underlie almost all physiological processes. The hyperLOPIT proteomics platform combines mass spectrometry with state-of-the-art machine learning to map the subcellular location of thousands of proteins simultaneously. We combine global proteome analysis with hyperLOPIT in a fully Bayesian framework to elucidate spatiotemporal proteomic changes during a lipopolysaccharide (LPS)-induced inflammatory response. We report a highly dynamic proteome in terms of both protein abundance and subcellular localisation, with alterations in the interferon response, endo-lysosomal system, plasma membrane reorganisation and cell migration. Proteins not previously associated with an LPS response were found to relocalise upon stimulation, the functional consequences of which are still unclear. By quantifying proteome-wide uncertainty through Bayesian modelling, a necessary role for protein relocalisation and the importance of taking a holistic overview of the LPS-driven immune response has been revealed. The data are showcased as an interactive application freely available for the scientific community.
Collapse
Affiliation(s)
- Claire M Mulvey
- Cambridge Centre for Proteomics, Cambridge Systems Biology Centre and Department of Biochemistry, University of Cambridge, Cambridge, CB2 1QR, UK
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, CB2 0RE, UK
| | - Lisa M Breckels
- Cambridge Centre for Proteomics, Cambridge Systems Biology Centre and Department of Biochemistry, University of Cambridge, Cambridge, CB2 1QR, UK
| | - Oliver M Crook
- Cambridge Centre for Proteomics, Cambridge Systems Biology Centre and Department of Biochemistry, University of Cambridge, Cambridge, CB2 1QR, UK
- MRC Biostatistics Unit, Cambridge Institute for Public Health, Forvie Site, Robinson Way, Cambridge, CB2 0SR, UK
| | - David J Sanders
- Department of Microbial Diseases, Eastman Dental Institute, University College London, Royal Free Campus, Rowland Hill Street, London, NW3 2PF, UK
| | - Andre L R Ribeiro
- Department of Microbial Diseases, Eastman Dental Institute, University College London, Royal Free Campus, Rowland Hill Street, London, NW3 2PF, UK
| | - Aikaterini Geladaki
- Cambridge Centre for Proteomics, Cambridge Systems Biology Centre and Department of Biochemistry, University of Cambridge, Cambridge, CB2 1QR, UK
| | | | - Nina Kočevar Britovšek
- Cambridge Centre for Proteomics, Cambridge Systems Biology Centre and Department of Biochemistry, University of Cambridge, Cambridge, CB2 1QR, UK
- Lek d.d., Kolodvorska 27, Mengeš, 1234, Slovenia
| | - Tracey Hurrell
- Cambridge Centre for Proteomics, Cambridge Systems Biology Centre and Department of Biochemistry, University of Cambridge, Cambridge, CB2 1QR, UK
| | - Michael J Deery
- Cambridge Centre for Proteomics, Cambridge Systems Biology Centre and Department of Biochemistry, University of Cambridge, Cambridge, CB2 1QR, UK
| | - Laurent Gatto
- Cambridge Centre for Proteomics, Cambridge Systems Biology Centre and Department of Biochemistry, University of Cambridge, Cambridge, CB2 1QR, UK
- de Duve Institute, UCLouvain, Avenue Hippocrate 75, Brussels, 1200, Belgium
| | - Andrew M Smith
- Department of Microbial Diseases, Eastman Dental Institute, University College London, Royal Free Campus, Rowland Hill Street, London, NW3 2PF, UK.
| | - Kathryn S Lilley
- Cambridge Centre for Proteomics, Cambridge Systems Biology Centre and Department of Biochemistry, University of Cambridge, Cambridge, CB2 1QR, UK.
| |
Collapse
|
148
|
Mizobuchi H, Soma GI. Low-dose lipopolysaccharide as an immune regulator for homeostasis maintenance in the central nervous system through transformation to neuroprotective microglia. Neural Regen Res 2021; 16:1928-1934. [PMID: 33642362 PMCID: PMC8343302 DOI: 10.4103/1673-5374.308067] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 10/26/2020] [Accepted: 12/16/2020] [Indexed: 12/25/2022] Open
Abstract
Microglia, which are tissue-resident macrophages in the brain, play a central role in the brain innate immunity and contribute to the maintenance of brain homeostasis. Lipopolysaccharide is a component of the outer membrane of gram-negative bacteria, and activates immune cells including microglia via Toll-like receptor 4 signaling. Lipopolysaccharide is generally known as an endotoxin, as administration of high-dose lipopolysaccharide induces potent systemic inflammation. Also, it has long been recognized that lipopolysaccharide exacerbates neuroinflammation. In contrast, our study revealed that oral administration of lipopolysaccharide ameliorates Alzheimer's disease pathology and suggested that neuroprotective microglia are involved in this phenomenon. Additionally, other recent studies have accumulated evidence demonstrating that controlled immune training with low-dose lipopolysaccharide prevents neuronal damage by transforming the microglia into a neuroprotective phenotype. Therefore, lipopolysaccharide may not a mere inflammatory inducer, but an immunomodulator that can lead to neuroprotective effects in the brain. In this review, we summarized current studies regarding neuroprotective microglia transformed by immune training with lipopolysaccharide. We state that microglia transformed by lipopolysaccharide preconditioning cannot simply be characterized by their general suppression of proinflammatory mediators and general promotion of anti-inflammatory mediators, but instead must be described by their complex profile comprising various molecules related to inflammatory regulation, phagocytosis, neuroprotection, anti-apoptosis, and antioxidation. In addition, microglial transformation seems to depend on the dose of lipopolysaccharide used during immune training. Immune training of neuroprotective microglia using low-dose lipopolysaccharide, especially through oral lipopolysaccharide administration, may represent an innovative prevention or treatment for neurological diseases; however more vigorous studies are still required to properly modulate these treatments.
Collapse
Affiliation(s)
- Haruka Mizobuchi
- Control of Innate Immunity, Technology Research Association, Kagawa, Japan
| | - Gen-Ichiro Soma
- Control of Innate Immunity, Technology Research Association, Kagawa, Japan
- Macrophi Inc., Kagawa, Japan
- Research Institute for Healthy Living, Niigata University of Pharmacy and Applied Life Sciences, Niigata, Japan
| |
Collapse
|
149
|
Bello-Corral L, Sánchez-Valdeón L, Casado-Verdejo I, Seco-Calvo JÁ, Antonio Fernández-Fernández J, Nélida Fernández-Martínez M. The Influence of Nutrition in Alzheimer's Disease: Neuroinflammation and the Microbiome vs. Transmissible Prion. Front Neurosci 2021; 15:677777. [PMID: 34489620 PMCID: PMC8417586 DOI: 10.3389/fnins.2021.677777] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 07/28/2021] [Indexed: 01/02/2023] Open
Abstract
Alzheimer's disease (AD) is a primary, progressive, neurodegenerative disorder. Many risk factors for the development of AD have been investigated, including nutrition. Although it has been proven that nutrition plays a role in AD, the precise mechanisms through which nutrition exerts its influence remain undefined. The object of this study is to address this issue by elucidating some of the mechanisms through which nutrition interacts with AD. This work is a qualitative systematic bibliographic review of the current literature searchable on various available databases, including PubMed, Web of Science, and Google Scholar. Our evidence comprises 31 articles selected after a systematic search process. Patients suffering with AD present a characteristic microbiome that promotes changes in microglia generating a proinflammatory state. Many similarities exist between AD and prion diseases, both in terms of symptoms and in the molecular mechanisms of pathogenesis. Changes in the composition of the gut microbiome due to dietary habits could be one of the environmental factors affecting the development of AD; however, this is probably not the only factor. Similarly, the mechanism for self-propagation of beta-amyloid seen in AD is similar to that seen in prions.
Collapse
Affiliation(s)
- Laura Bello-Corral
- Department of Nursing and Physical Therapy, University of León, León, Spain
| | | | | | - Jesús Ángel Seco-Calvo
- Institute of Biomedicine, University of León, León, Spain.,University of the Basque Country, Leioa, Spain
| | | | | |
Collapse
|
150
|
Tualang Honey: A Decade of Neurological Research. Molecules 2021; 26:molecules26175424. [PMID: 34500857 PMCID: PMC8434576 DOI: 10.3390/molecules26175424] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 09/01/2021] [Accepted: 09/02/2021] [Indexed: 02/06/2023] Open
Abstract
Tualang honey has been shown to protect against neurodegeneration, leading to improved memory/learning as well as mood. In addition, studies have also demonstrated its anti-inflammatory and antioxidant properties. However, a substantial part of this research lacks systematization, and there seems to be a tendency to start anew with every study. This review presents a decade of research on Tualang honey with a particular interest in the underlying mechanisms related to its effects on the central nervous system. A total of 28 original articles published between 2011 and 2020 addressing the central nervous system (CNS) effects of Tualang honey were analysed. We identified five main categories, namely nootropic, antinociceptive, stress-relieving, antidepressant, and anxiolytic effects of Tualang honey, and proposed the underlying mechanisms. The findings from this review may potentially be beneficial towards developing new therapeutic roles for Tualang honey and help in determining how best to benefit from this brain supplement.
Collapse
|